WO2018163216A1 - Spirooxindole compounds as gsk3β inhibitors and process for preparation thereof - Google Patents

Spirooxindole compounds as gsk3β inhibitors and process for preparation thereof Download PDF

Info

Publication number
WO2018163216A1
WO2018163216A1 PCT/IN2018/050134 IN2018050134W WO2018163216A1 WO 2018163216 A1 WO2018163216 A1 WO 2018163216A1 IN 2018050134 W IN2018050134 W IN 2018050134W WO 2018163216 A1 WO2018163216 A1 WO 2018163216A1
Authority
WO
WIPO (PCT)
Prior art keywords
compounds
spiro
methyl
triazol
furan
Prior art date
Application number
PCT/IN2018/050134
Other languages
French (fr)
Other versions
WO2018163216A9 (en
Inventor
Prathama S MAINKAR
Mohammad Abdul SATTAR
Pitchakuntla MALLESHAM
Togapur Pavan Kumar
Divya DUSCHARLA
Ummanni RAMESH
Srivari CHANDRASEKHAR
Original Assignee
Council Of Scientific & Industrial Research
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Council Of Scientific & Industrial Research filed Critical Council Of Scientific & Industrial Research
Priority to EP18715984.3A priority Critical patent/EP3592747A1/en
Priority to US16/492,359 priority patent/US11306100B2/en
Publication of WO2018163216A1 publication Critical patent/WO2018163216A1/en
Publication of WO2018163216A9 publication Critical patent/WO2018163216A9/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to spirooxindole compounds useful as GSK3P inhibitors and process for preparation thereof. Particularly the present invention relates to spirooxindole compounds of general formula I.
  • Z is hydrogen, halogen, hydroxy, alkoxy, cyano, nitro
  • Ring A is a five membered or six membered or seven membered optionally containing additional hetero atom in the ring
  • X is optionally C3 ⁇ 4
  • Certain spirooxindoles were also found to exhibit good analgesic and anesthetic properties (J. Med. Chem. 1976, 19, 892) and are particularly effective in treatment of chronic pain (US Pat. No. 6774132). Spirooxindole compounds are also used as aldose reductase inhibitors and are used in the treatment of diabetes. It is not surprising that these compounds have been the focus of many chemists, in part because of their biological activities, but also the challenge of the simultaneous creation of spiro-quaternary centers and multiple chiral centers. As a result, numerous elegant transformations have been developed for the effective construction of these structural skeletons (Adv. Synth. Catal. 2013, 355, 1023; Org. Lett.
  • Z is hydrogen, halogen, hydroxy, alkoxy, cyano, nitro
  • Ring A is a five membered or six membered or seven membered optionally containing additional hetero atom in the ring
  • X is optionally CH 2 or none
  • Ring B is triazole or cyclic amine alone or with one or more hetero atoms part of the ring and optionally substituted with one or more substituents or no B ring present
  • Y is hydrogen, halogen, azide, alkoxy, aryloxy, hydroxy, ester, amide, amino, alkyl or aryl, heteroaryl, wherein these substituents are optionally substituted with one or more halogen, hydroxy, alkoxy, alkyl, aryl, heteroaryl, aryloxy, nitro, cyano, ester, aldehyde,
  • the present invention provides new class of spirooxindoles which were synthesized from oxindole.
  • kinase reaction mixture was prepared by adding 5 mg of recombinant enzyme, 2 ⁇ ATP, 1 ⁇ g of enzyme substrate along with test compound or DMSO to a final volume of 5 ⁇ . The kinase reaction was performed by incubating the mixture at room temperature for 17 mins. To correct reaction back ground a substrate blank reaction was also performed.
  • the cells were fixed using 10% TCA and incubated for 1 h at 4 °C. Then the plates were rinsed carefully with MQ water and air dried at room temperature. To all the wells of MTP plate 0.057% SRB solution was added and kept for approximately 30 min before it was rinsed off using 1% acetic acid. The plates were then air dried and 100 ⁇ ⁇ of 10 mM tris base was added to each well to solubilize the SRB before absorbance was measured at 510 nm. The measure of absorbance is directly proportional to cell growth and is thus used to calculate % reduction in the rate of cell proliferation according to control DMSO treated cells. All experiments were performed in triplicates; mean % age proliferation values with + SD were plotted here.
  • TRAIL induced apoptosis of cancer cell is mediated by the activation of procaspases to active caspases by proteolysis.
  • the compounds show synergistic effect on activation of caspase cascade to induce apoptosis, we have measured caspase-3 and -9 activities in PANC- 1 cells treated with compounds along with TRAIL.
  • the obtained result suggests that the combinatorial treatment of PANC- 1 cells is more effective even at lower concentration of the compounds than individually in activation of both the caspases ( Figure 5).
  • the apoptosis of PANC-1 cells treated with GSK3P inhibitors is through mitochondiral dependant pathway.
  • GSKP participates in regulation of wnt pathway via phosphorylation of ⁇ -catenin. Active GSK3P phosphorylates ⁇ -catenin targeting it for ubiquitin mediated proteolysis. Inhibition of GSK3P causes the stabilization and accumulation of ⁇ -catenin in cytoplasm and further translocates into the nucleus. Nuclear ⁇ catenin along with TCF/LEF elements continuously drive transcription of its target genes. Nuclear ⁇ -catenin is implicated in progression of several types of cancers such as prostate cancer, basal cell carcinoma, head and neck cancers and medulloblastoma.
  • Glycogen Synthase Kinase 3 ⁇ is a serine/threonine kinase. It is a multi-functional enzyme, regulating several proteins involved in various signalling pathways including transcription factors, cell cycle/survival regulators and proto-oncogenes. Recent studies suggest that GSK3P may promote turn oogenesis in many cancer types. Its abberent over expression was found in human pancreatic (H), colon(I), prostate(J), leukaemia (K), thyroid(l), brain(M) and ovarian(N) cancer cells. Billadeau et. al. reported for elevated levels of GSK3P in many pancreatic cancer cell lines including PANC-1.
  • nfkb positively regulates the transcription of anti-apoptotic proteins such as XIAP, BCL-2, BCL- XL, and MCL-1 in cancer cells
  • GSK mediates binding NFKB P65 subunit to the promoters of an anti-apoptotic genes BCL2 and XIAP, so inhibition of GSK reduces the expression these anti-apoptotic genes, there by inhibition of GSK sensitize the pancreatic cancer cells to trail mediated apoptosis.
  • Another advantage of the present invention is the use of these spirooxindole compounds as GSK3P inhibitors.

Abstract

The present invention provides a compound of general formulae I useful as GSK3β inhibitors and their anticancer potential and process for preparation of these spirooxindoles. Formula I wherein Z is hydrogen, halogen, hydroxy, alkoxy, cyano, nitro; Ring A is a five membered or six membered or seven membered optionally containing additional hetero atom in the ring X is optionally CH2 Ring B is triazole or cyclic amine alone or with one or more hetero atoms part of the ring and optionally substituted with one or more substituents Y is hydrogen, alkoxy, aryloxy, hydroxy, ester, amide, amino, alkyl or aryl, heteroaryl, wherein these substituents are optionally substituted with one or more halogen, hydroxy, alkoxy, alkyl, aryl, heteroaryl, aryloxy, nitro, cyano, ester, aldehyde, wherein these substituents are further substituted with one or more halogen, hydroxy, alkoxy, alkyl, aryl, heteroaryl, aryloxy, nitro, cyano, ester, aldehyde.

Description

SPIROOXINDOLE COMPOUNDS AS GSK3P INHIBITORS AND PROCESS FOR
PREPARATION THEREOF FIELD OF THE INVENTION
The present invention relates to spirooxindole compounds useful as GSK3P inhibitors and process for preparation thereof. Particularly the present invention relates to spirooxindole compounds of general formula I.
Figure imgf000003_0001
Formula I
wherein
Z is hydrogen, halogen, hydroxy, alkoxy, cyano, nitro;
Ring A is a five membered or six membered or seven membered optionally containing additional hetero atom in the ring
X is optionally C¾
Ring B is triazole or cyclic amine alone or with one or more hetero atoms part of the ring and optionally substituted with one or more substituents
Y is hydrogen, alkoxy, aryloxy, hydroxy, ester, amide, amino, alkyl or aryl, heteroaryl, wherein these substituents are optionally substituted with one or more halogen, hydroxy, alkoxy, alkyl, aryl, heteroaryl, aryloxy, nitro, cyano, ester, aldehyde,
wherein these substituents are further substituted with one or more halogen, hydroxy, alkoxy, alkyl, aryl, heteroaryl, aryloxy, nitro, cyano, ester, aldehyde;
BACKGROUND OF THE INVENTION
Oxindoles substituted in the 3 position are an important structural motif among a series of natural substances and pharmaceutically effective substances. Some of these compounds exhibit biological activity against various pathogens and have e.g. antitumor or anti-HIV properties (J. Med. Chem. 2006, 49, 3432; Bioorg. Med. Chem. Lett. 2006, 16, 2105). Among the 3-substituted oxindoles, spirocyclic oxindoles are the structural motifs frequently found in many natural products and biologically active molecules. The key structural characteristic of these compounds is the spiro ring fused at the 3 -position of the oxindole core, with varying degrees of substitution around it. The alkaloids such as mitraphylline, pteropodine, gelsemine, speciophylline, uncarine, Strychnofoline, Alstonisine, Rhychnophylline, Spirotriprostatin A, Salacin, Elocamine, Horsifiline, Coerulesine etc., are some of the example of natural products containing spirooxindole structural framework. Spirooxindole derivatives were found to exhibit good biological properties (PCT Patent Application No. WO 2006/110917, WO 2010/45251, WO 2014/058035, WO 2011/047174; PCT/US2010/040187; EP 2488531, US 8101647, US 8742109, US 6358750, US 8962828). These are useful for the treatment of sodium-channel mediated diseases, diseases related to pain, central nervous conditions (such as epilepsy, anxiety, depression and bipolar disease), cardiovascular conditions (such as arrhythmias, atrial fibrillation and ventricular fibrillation), neuromuscular conditions such as (restless syndrome, neuroprotection against stroke, neural trauma, and multiple sclerosis), channelopathesis such as erythromelalgia and familial rectal pain syndrome. Certain spirooxindoles were also found to exhibit good analgesic and anesthetic properties (J. Med. Chem. 1976, 19, 892) and are particularly effective in treatment of chronic pain (US Pat. No. 6774132). Spirooxindole compounds are also used as aldose reductase inhibitors and are used in the treatment of diabetes. It is not surprising that these compounds have been the focus of many chemists, in part because of their biological activities, but also the challenge of the simultaneous creation of spiro-quaternary centers and multiple chiral centers. As a result, numerous elegant transformations have been developed for the effective construction of these structural skeletons (Adv. Synth. Catal. 2013, 355, 1023; Org. Lett. 2011, 13, 3086; Angew. Chem. 2004, 116, 2719). Although many types of spirooxindole derivatives were reported using a variety of strategies toward the construction of spirooxindole architecture, there are certain groups of spirooxindole architecture of interest that have not been synthesized and evaluated for biological properties. Spirooxindole- triazoles and spirooxindole-cyclic amines are examples of this kind and are impossible to find naturally. Therefore, there is a need for the development of methods for the synthesis and biological evaluation of diversely substituted spirooxindoles containing triazole and cyclic amine motifs and others.
In this context a large number of new spirooxindoles architectures possessing triazole and cyclic amines have been synthesized and evaluated for GSK3P activity and their anticancer potential.
OBJECTIVE OF THE INVENTION
The main objective of the present invention is to provide novel spirooxindole compounds as useful GSK3P inhibitors.
Another objective of the present invention is to provide the process for the preparation of novel spirooxindoles compounds.
SUMMARY OF THE INVENTION
The above and other objectives of the present invention are achieved by providing the new spirooxindole compounds, which have been synthesized from oxindole.
Accordingly, the present invention affords a new class of spirooxindole of general formula I.
Figure imgf000005_0001
Formula I
wherein
Z is hydrogen, halogen, hydroxy, alkoxy, cyano, nitro;
Ring A is a five membered or six membered or seven membered optionally containing additional hetero atom in the ring
X is optionally CH2 or none
Ring B is triazole or cyclic amine alone or with one or more hetero atoms part of the ring and optionally substituted with one or more substituents or no B ring present
Y is hydrogen, halogen, azide, alkoxy, aryloxy, hydroxy, ester, amide, amino, alkyl or aryl, heteroaryl, wherein these substituents are optionally substituted with one or more halogen, hydroxy, alkoxy, alkyl, aryl, heteroaryl, aryloxy, nitro, cyano, ester, aldehyde,
wherein these substituents are further substituted with one or more halogen, hydroxy, alkoxy, alkyl, aryl, heteroaryl, aryloxy, nitro, cyano, ester, aldehyde;
The structural formulas of the representative compounds are
Figure imgf000006_0001
Figure imgf000006_0002
Figure imgf000006_0003
Figure imgf000006_0004
8
Formula III wherein the example of specific compounds are as follows
Figure imgf000007_0001
Figure imgf000007_0002
wherein the example of specific compounds are as follow
Figure imgf000008_0001
AC-J AC-K AC-L
Structures of the other intermediated compounds in the process for the preparation of spirooxindole derivatives are represented as
Figure imgf000008_0002
6a 6b 7a 7b In an embodiment of the present invention, the novel spirooxindole derivatives described herein are represented by
(AC-A) Ethyl 6-(5-fluoro-2-((l-(2-oxo-2', 4', 5', 6'-tetrahydrospiro[indoline-3,3'-pyran]-5'- yl)- 1 H- 1 ,2,3-triazol-4-yl)methoxy)-phenyl)-2-methylnicotinate
(AC-B) Ethyl 2-methyl-6-(2-((l-(2-oxo-2',4',5',6'-tetrahydrospiro[indoline-3,3'-pyran]-5'-yl)- lH-l,2,3-triazol-4-yl)methoxy)-phenyl)nicotinate
(AC-C) Ethyl 6-(5-chloro-2-((l-(2-oxo-2',4',5',6'-tetrahydrospiro [indoline-3,3'-pyran]-5'-yl)- lH-l,2,3-triazol-4- yl)methoxy)-phenyl)- 2-methyl nicotinate
(AC-D) Ethyl 6-(3,5-dichloro-2-((l-(2-oxo-2',4',5',6'-tetrahydrospiro-[indoline-3,3'-pyran]-5'- yl)- 1 H- 1 ,2,3-triazol-4-yl)methoxy)phenyl)-2-methyl nicotinate
(AC-E) 2-((l-(2-oxo-2', 4', 5', 6'-tetrahydrospiro[indoline-3,3'-pyran]-5'-yl)-lH-l,2,3-triazol- 4-yl) methoxy)benzaldehyde
(AC-F) 5'-(4-(pyridin-2-yl)-lH-l,2,3-triazol-l-yl)-2',4',5',6'-tetrahydrospiro[indoline-3,3'- pyran]-2-one
(AC-G) 5'-(4-(hydroxy(phenyl)methyl)-lH-l,2,3-triazol-l-yl)-2',4',5',6'-tetrahydrospiro [indoline-3 ,3 '-pyran] -2-one
(AC-H) 5'-(4-((2-nitrophenoxy)methyl)-lH-l,2,3-triazol-l-yl)-2',4',5',6'-tetrahydrospiro - [indoline-3 ,3 '-pyran] -2-one
(AC-I) 5'-(4-((4-chloro-2-methylphenoxy)methyl)-lH-l,2,3-triazol-l-yl)-2',4',5',6'-tetra hydrospiro[indoline-3,3'-pyran]-2-one
(AC-J) 5'-(4-((3-hydroxyphenylamino)methyl)-lH-l,2,3-triazol-l-yl)-2',4',5',6'-tetrahydro spiro [indoline-3 ,3 '-pyran] -2-one
(AC-K) 5'-(4-((3-aminophenoxy)methyl)-lH-l,2,3-triazol-l-yl)-2',4',5',6'-tetrahydro spiro [indoline-3 ,3 '-pyran] -2-one
(AC-L) 5'-(4-((5-bromo-3-nitropyridin-2-yloxy) methyl)-lH-l,2,3-triazol-l-yl)-2',4',5',6'- tetrahydrospiro[indoline-3,3'-pyran]-2-one
(AB-O) Ethyl 2-methyl-6-(2-((l-((2'-oxo-4,5-dihydro-2H-spiro[furan-3,3'-indoline]-5- yl)methyl)- 1H- 1 ,2,3-triazol-4-yl)methoxy)phenyl)nicotinate
(AB-M) Ethyl 6-(5-fluoro-2-((l-((2'-oxo-4,5-dihydro-2H-spiro[furan-3,3'-indoline]-5- yl)methyl)- 1H- 1 ,2,3-triazol-4-yl)methoxy)phenyl)-2-methylnicotinate
(AB-N) Methyl-6-(5-chloro-2-((l-((2'-oxo-4,5-dihydro-2H-spiro[furan-3,3'-indoline]-5- yl)methyl)- 1H- 1 ,2,3-triazol-4-yl)methoxy)phenyl)-2-methylnicotinate (AB-P) 2-((l-((2'-oxo-4,5-dihydro-2H-spiro[furan-3,3'-indoline]-5-yl)methyl)-lH-l,2,3- triazol-4-yl)methoxy)benzaldehyde
(AB-W) Ethyl6-(3,5-dichloro-2-((l-((2'-oxo-4,5-dihydro-2H-spiro[furan-3,3'-indoline]-5- yl)methyl)- 1H- 1 ,2,3-triazol-4-yl)methoxy)phenyl)-2-methylnicotinate
(AB-F) Ethyl-l-((2'-oxo-4,5-dihydro-2H-spiro[furan-3,3'-indoline]-5-yl)methyl)-lH-l,2,3- triazole-4-carboxylate
(AB-C) 5-((4-phenyl H-l,2,3-triazol-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]- 2'-one
(AB-G) 5-((4-(hydroxymethyl)-lH-l,2,3-triazol-l-yl)methyl)-4,5-dihydro-2H-spiro[furan- 3,3'-indolin]-2'-one
(AB-E) 5-((4-(2-hydroxypropan-2-yl)-lH-l,2,3-triazol-l-yl)methyl)-4,5-dihydro-2H- spiro [furan-3 ,3 '-indolin] -2'-one
(AB-D) 5-((4-(4-nitrophenyl)-lH-l,2,3-triazol-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'- indolin]-2'-one
(AB-X) 5-((4-phenylpiperazin-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]-2'-one (AB-K) 5-(morpholinomethyl)-4, 5-dihydro-2H-spiro [furan-3, 3' -indolin] -2'-one
(AB-S) 5-((4-(pyridin-2-yl)piperazin-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]- 2'-one
(AB-U) 5-((4-(2-methoxyphenyl)piperazin-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'- indolin] -2'-one
(AB-T) 5-((4-(4-methoxyphenyl)piperazin-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'- indolin] -2'-one
(AB-Q) 5-((4-benzoylpiperazin-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]-2'-one (AB-L) 5-(thiomorpholinomethyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]-2'-one
(AB-R) 5-((4-benzylpiperazin-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]-2'-one (AB-V) 5-((4-(bis(4-fluorophenyl)methyl)piperazin-l-yl)methyl)-4,5-dihydro-2H-spiro [furan-3 ,3 '-indolin] -2'-one
(6a) 5-(iodomethyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]-2'-one
(6b) 5'-Iodo-2', 4', 5', 6'-tetrahydrospiro [indoline-3,3'-pyran]-2-one
(7a) 5-(azidomethyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]-2'-one
(7b) 5'-azido-2',4',5',6'-tetrahydrospiro[indoline-3,3'-pyran]-2-one
The present invention also provides a process for the preparation of spirooxindole compounds as described in the above formula A large number of various spirooxindoles possessing diversely substituted architecture were found to exhibit several biological properties. These functionalities are prominent structural motifs of new medicines from different pharmacological groups. The development of new structural scaffolds of spirooxindole architecture is very important for the drug discovery process. In this connection a large number of spirooxindole derivatives were developed as depicted in the above general formula I to IV.
The process for the synthesis of these new spirooxindoles starts from the allylation of oxindole and involves operationally simple and highly efficient synthetic protocol giving rise to the desired products in high yields.
In an embodiment of the present invention commercially available oxindole was used as the starting material for the complete process.
In another embodiment of the present invention the first step is allylation of oxindole
In another embodiment of the present invention the second step is protection of the free amine with Boc group.
In yet another embodiment of the present invention the third step is introduction of primary hydroxyl using formaldehyde
In yet another embodiment of the present invention the fourth step is removal of the protection on free amine.
In yet another embodiment of the present invention the fifth step is cyclisation using iodine to give spiroiodo compounds.
In yet another embodiment of the present invention the sixth step is conversion of iodo to azide to give spiroazide compounds.
In still another embodiment of the present invention the seventh step is treatment of spiroazides with alkynes to give triazole based spirooxindoles.
In still another embodiment of the present invention the reaction of spiroiodo compounds with cyclic amines gives the cyclic amine based spirooxindoles.
In still another embodiment of the present invention the spirooxindole compounds prepared are tested for their efficiency towards GSK3P inhibition property and their anticancer potential.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1: Screening of GSK3P Inhibitors Using the ADP-Glo Kinase Assay:
Figure 2: GSK3P inhibition sensitizes pancreatic cancer cells (PANC-1) to TRAIL:
Figure 3: (A) The cytotoxicity results indicate that the compounds significantly increased sensitivity of PANC-1 cells to TRAIL; (B) The compound AB-C is not effective compared to remaining compounds
Figure 4: The TRAIL alone did not show effective inhibition on colony formation at the concentration 10 ng/mL used
Figure 5: Activity AB-0 and AB-U on cellular GSK3P signalling:
Figure 7: Effect of AB-0 and AB-U on expression of NFkB target genes involved in cell survival:
Figure 8: molecules AB-0 and AB-U inhibiting cellular GSK3P sensitizing the PANC-1 to TRAIL inducing apoptosis.
DETAILED DESCRIPTION OF THE INVENTION
Spirooxindoles are efficient structural motifs capable of showing diverse biological activities. This resulted in design and synthesis of a large number of spirooxindoles as illustrated in Scheme 1, which comprises of:
The process for the preparation of spirooxindoles wherein the said process comprising the steps of:
a) allylation of oxindole
b) protection of free amine with boc group
c) formylation using formaldehyde
d) removal of the protection to give free amine
e) cyclisation using iodine to give spiro-iodo compounds
f) azidation of spiro-iodo compounds using sodium azide to give spiro-azide compounds or reaction of spiro-iodo compounds with cyclic amines to give the cyclic amine based spirooxindoles
g) reaction of spiro-azide compounds with alkynes to give triazole based spirooxindoles under click chemistry conditions.
EXAMPLES
The present invention will be more specifically explained by following examples. However, the scope of the present invention is not limited to the scope of the examples stated below. Experimental section
Example 1
3-Allylindolin-2-one (2)
To a stirred solution of oxindole (10g,75 mmol) in dry THF (200 mL) under inert condition was added tetra methyl ethylene diamine (TMEDA) (22.5g,150 mmol) and the resulting solution was cooled to -78 °C. After 10 min, nBuLi (60 mL, 2.5 M in hexanes) was added slowly drop by drop over 30 min, and then allowed to stir for another 30 min. Then allyl bromide (16g, 124.34 mmol) was added slowly to the reaction mixture, warmed to -23 °C, and maintained for 4h, after the completion of reaction (monitored by TLC) saturated NH4C1 (100 mL) was added and extracted with EtOAc (3x200 mL). The combined organic layers were washed with brine and dried over Na2S04. The solvent was evaporated in vacuum and the residue was purified by column chromatography on silica gel eluting with hexanes/EtOAc (70:30) to give allyl oxindole (9.07g, 70% yield).
1H NMR (300 MHz, CDC13): δ 8.71 (br s, 1H), 7.27 (d, 1 H, = 5.7 Hz), 7.21 (t, 1H, = 7.6 Hz), 7.02 (t, 1H, = 7.6 Hz), 7.02 (td, 1H, = 6.8 Hz), 6.90 (td, 1H, = 7.7 Hz), 5.77 (m, 1H), 5.18-5.04 (dd, 2H, J = 10.0 Hz), 3.53 (q, 1H, J = 5.0 Hz), 2.84 (m, 1H), 2.60 (m, 1H); 13C NMR (300MHz, CDC13): δ 180.1, 141.2, 133.8, 129.2, 127.2, 124.3, 122.1, 118.0, 109.8, 45.7, 34.7; ESI-MS: m/z 174 [M+H]+.
Example 2
ie/t-Butyl 3-allyl-2-oxoindoline-l-carboxylate (3)
To a stirred solution of allyl oxindole 2 (5g, 28.9 mmol) in dry THF (50 mL), was added Na2C03 (6.123 g, 57.08 mmol) at room temperature, after 5 min (Boc)20 (6.9g, 31.97 mmol) was added and then reaction mixture was heated to 60 °C. The reaction was allowed to stir vigorously for 6 h under N2-atmosphere. Then the reaction mixture was cooled to room temperature, diluted with water and extracted with EtOAc (2x100 mL). The combined organic layers were given a brine wash (20 mL) and dried over anhydrous Na2S04. The solvent was concentrated in vacuo and the residue was purified by silica gel column using EtOAc/petroleum ether (5:95) to afford the Boc protected allyl oxindole compound 3 (6.39 g, 80% yield).
1H NMR (300 MHz, CDC13): δ 7.80 (d, 1H, = 8.1 Hz), 7.34-7.25 (m, 2H), 7.14 (t, 1H, = 7.3 Hz), 5.83-5.67 (m, 1H), 5.18-5.01 (m, 2H), 3.61 (t, 1H, J = 6.0 Hz), 2.89-2.77 (m, 1H), 2.71-2.51 (m, 1H), 1.64 (s, 9H); 13C NMR (75 MHz, CDC13): δ 175.5, 149.1, 133.4, 131.5, 130.8, 128.0, 124.1, 123.9, 118.5, 114.8, 84.2, 45.6, 35.4, 27.9; MASS (ESI): m/z 296 [M+Na]+.
Example 3
ie/t-Butyl 3-allyl-3-(hydroxymethyl)-2-oxoindoline -1-carboxylate (4)
To a stirred solution of Boc oxindole 3 (5.0g, 18.30 mmol) in dry DME (30 mL) was added quinine (0.059 g, 0.183 mmol), followed by HCHO (0.82g, 27.84 mmol) and the resulting mixture was stirred at room temperature for 20 h. After completion of the reaction (monitored by TLC), the reaction mixture was partitioned between water (20 mL) and EtOAc (50 mL). The organic layer was separated and the aqueous phase was extracted with EtOAc (2x40 mL) and the combined organic layers were washed with brine and dried over Na2S04.The solvent was evaporated in vacuo and the residue was purified by column chromatography on silica gel eluted with hexanes/EtOAc (85:15) to give hydroxyl methyl compound 4 (5.43 g, 98% yield).
1H NMR (300 MHz, CDC13): δ 7.80 (d, 1H, = 8.1 Hz), 7.34-7.25 (m, 2H), 7.14 (t, 1H, = 7.3 Hz), 5.83-5.67 (m, 1H), 5.18-5.01 (m, 2H), 4.15-3.92 (m, 2H), 2.89-2.77 (m, 1H), 2.71- 2.51 (m, 1H), 1.54 (s, 9H); 13C NMR (75 MHz, CDC13): δ 175.9, 151.5, 149.0, 135.4, 130.8, 128.0, 124.1, 123.9, 118.5, 114.8, 84.2, 79.2, 45.6, 35.4, 27.9; MASS (ESI): m/z 326 [M+Na]+.
Example 4
3-Allyl-3-(hydroxymethyl) indolin-2-one (5)
To a solution of alcohol 4 (2.0g, 6.60 mmol) in dichloromethane (30 mL) was added trifluoroacetic acid (1.52g, 13.76 mmol) at 0 °C. The reaction mixture was stirred at 0 °C for lh and another 6h at room temperature. After completion of the reaction (monitored by TLC), the solvent and TFA was removed under reduced pressure below 45 °C. The residue was diluted with CH2C12 (50 mL) and water (50 mL). The organic layer was separated, washed with brine (20 mL), the combined organic layer was dried over anhydrous Na2S04 and concentrated in vacuo. The residue was purified by silica gel column chromatography using petroleum ether/EtOAc (75:25) to afford Boc deprotected compound 5 (1.07 g, 80% yield) as yellowish liquid.
1H NMR (300 MHz, CDC13): δ 8.18 (s, 1H), 7.27 (m, 2H), 7.07 (m, 1H), 6.90 (d, = 2.7 Hz, 1H), 5.49 (m, 1H), 5.04 (m, 1H), 4.95 (dd, J = 10.2 Hz, 1H), 3.92 (d, J = 11.2 Hz, 1H), 3.80 (d, J = 10.9 Hz, 1H), 2.64 (m, 2H), 1.72 (s, 1H); 13C NMR (75MHz, CDC13): δ 180.0, 141.5, 133.8, 129.1, 127.9, 124.3, 122.1, 118.0, 109.7, 66.4, 54.9, 37.2; ESI-MS: m/z 204 [M+H]+; IR (Neat): 3437, 2982, 1789, 1730, 1609, 1152, 844, 771, 676 cm"1.
General Procedure for Spiro-Iodo compounds (6a and 6b)
To the solution alcohol 5 (2g, 9.85 mmol) in acetonitrile (30 mL), were added iodine (5.52g, 20.68 mmol) and Na2C03 (2.19g, 20.68 mmol), and the resulting reaction mixture was stirred for 12 h at reflux. After complete conversion of starting material (monitored by TLC), reaction was quenched with saturated hypo solution (20 mL), then stirred for another 15 min, and extracted with EtOAc (3x50 mL). Combined organic layers were washed with brine (50 mL), dried over anhydrous Na2S04 and concentrated in vacuo. The residue was purified by silica gel column chromatography using petroleum ether/EtOAc (80:20) to afford spiro-iodo compounds
Example 5
5-(Iodomethyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]-2'-one (6a)
Yield = 78%; White solid; mp-170.6 °C; IR (neat): vmax 3243, 2927, 1711, 1619, 1341, 1188, 1070, 752, 632 cm"1; 1H NMR (300 MHz, CDC13): δ 8.48 (br, s, 1H), 7.21-7.28 (m, 2H), 7.09 (t, = 7.5 Hz, 1H), 6.95 (d, = 8.3 Hz, 1H), 4.57-4.63 (m, 1H), 4.30 (d, = 8.3 Hz, 1H), 4.01 (d, = 8.3 Hz, 1H), 3.41-3.47 (m, 2H), 2.35-2.41 (m, 2H); 13CNMR (75 MHz, CDC13): δ 180.2, 140.0, 133.3, 128.3, 123.0, 122.8, 110.2, 80.2, 76.3, 55.8, 44.1, 8.0; ESI-MS: m/z 352 [M+Na]+; HRMS: calcd for Ci2Hi2I02NNa 352.1110 found 352.1252.
Example 6
5'-Iodo-2', 4', 5', 6'-tetrahydrospiro [indoline-3,3'-pyran]-2-one (6b)
3.17g 78% yield); white solid.
1H NMR (300 MHz, CDC13): δ Ί .39 (d, 1H, = 7.4 Hz), 7.27-7.21 (m, 2H), 7.08 (t, 1H, = 7.4 Hz), 6.93 (d, 1H), 4.39-4.32 (m, 1H), 4.16 (s, 2H), 3.52-3.42 (m, 2H), 2.63 (dd, 1H, =
6.4 Hz & 13.1 Hz), 2.04 (dd, 1H, = 9.0 Hz & 12.9 Hz); 13C NMR (75 MHz, CDC13): δ 181.0, 140.2, 133.0, 128.3, 123.1, 123.1, 109.8, 79.1, 77.0, 55.2, 44.6, 8.3; MASS (ESI): m/z 252 [M+Na]+.
Example 7
5-(Azidomethyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]-2'-one (7a)
To a solution of iodide 6a (2.5g, 7.58 mmol) in dry DMF (15 mL) was added NaN (0.74g, 11.3 mmol) under inert condition and the resulting mixture was heated to 80 °C for 12 h. After completion of the reaction (monitored by TLC), reaction mixture was diluted with cool water (30 mL), and extracted with diethyl ether (2x75 mL). Organic layer was washed with cool water for two times and the layers are separated. The organic layer was washed with brine (20 mL), dried over anhydrous Na2S04 and concentrated in vacuo. The crude azide obtained was purified by column chromatography on silica gel eluted with hexanes/EtOAc (80:20) to give spiro-azide 7a (2.0g, 81% yield) as a pale yellow liquid.
Yield = 81%; liquid; IR (neat): vmax 3247, 2925, 2856, 2101, 1712, 1619, 1471, 1082, 752, 681 cm"1; 1H NMR (300 MHz, CDC13): δ 8.35 (br, s, 1H), 7.37 (d, = 7.5Hz,lH), 7.26 (t, =
7.5 Hz, 1H), 7.08 (t, J = 7.5 Hz, 1H), 6.90 (d, = 6.9 Hz, 1H), 4.51-4.58 (m, 1H), 4.1 (s, 2H), 3.68 (dd, = 3.7 Hz,1.2Hz 1H), 3.42 (dd, = 5.2 Hz, 3.2 Hz, 1H), 2.52 (dd, = 6.7 Hz,5.1Hz 1H), 2.12 (dd, = 9.8 Hz, 3.6Hz, 1H); 13C NMR (75 MHz, CDC13): δ 180.1, 141.1, 132.3, 128.3, 122.0, 121.8, 111.2, 80.2, 75.3, 56.8, 55.1, 37.2; ESI-MS: m/z 245 [M+H]+; HRMS: calcd for Ci2Hi202N 4Na 247.1210 found 247.1252.
Example 8
5'-Azido-2',4',5',6'-tetrahydrospiro[indoline-3,3'-pyran]-2-one (7b)
To a solution of iodide 6b (2.5g, 7.58 mmol) in dry DMF (15 mL) was added NaN3 (0.74g, 11.3 mmol) under inert condition and the resulting mixture was heated to 80 °C for 12 h. After completion of the reaction (monitored by TLC), reaction mixture was diluted with cool water (30 mL), and extracted with diethyl ether (2x75 mL). Organic layer was washed with cool water for two times and the layers are separated. The organic layer was washed with brine (20 mL), dried over anhydrous Na2S04 and concentrated in vacuo. The crude azide obtained was purified by column chromatography on silica gel eluted with hexanes/EtOAc (80:20) to give spiro-azide 7b (2.0g, 81% yield) as a pale yellow liquid.
1H NMR (300 MHz, CDC13): δ 9.28 (br, s, 1H), 7.35 (d, 1H, = 7.3 Hz), 7.24 (td, 1H, = 7.7 Hz, 1.2 Hz), 7.09 (td, 1H, = 7.6 Hz, 0.9 Hz), 6.95 (d, 1H, = 7.7 Hz), 4.58-4.53 (m, 1H), 4.11 (s, 2H), 3.67 (dd, 1H, = 12.9 Hz, 3.6 Hz), 3.46 (dd, 1H, = 13.1 Hz, 5.3 Hz), 2.51 (dd, 1H, = 12.8 Hz & 6.7 Hz), 2.12 (dd, 1H, = 12.9 Hz , 3.6 Hz); 13C NMR (75 MHz, CDC13): δ 180.0, 140.0, 133.3, 128.3, 123.0, 122.8, 110.2, 79.1, 76.1, 55.2, 53.8, 41.0; MASS (ESI): m/z 267 [M+Na]+.
General procedure for spirooxindole-triazoles (9a and 9b)
Spiro-azide 7a/7b (0.5 mmol), terminal alkyne (0.5 mmol), dry Et3N (1.0 mmol), THF (6 mL) and dry Cul (0.5 mmol) were added successively under N2 atmosphere. Reaction mixture was stirred for 2 h at room temperature, and then water (3 mL) was added, crude was extracted with EtOAc (3x15 mL). The combined organic layers were washed with brine solution and dried over Na2S04. The solvent was evaporated in vacuo and the residue was purified by column chromatography to afford triazole (9a and 9b).
Example 9
Ethyl 6-(5-fluoro-2-((l-(2-oxo-2', 4', 5', 6'-tetrahydrospiro[indoline-3,3'-pyran]-5'-yl)-lH- 1 ,2,3-triazol-4-yl)methoxy)-phenyl)-2-methylnicotinate (AC-A)
1H NMR (300 MHz, CDC13): δ 8.17 (d, 1H, = 8.2 Hz), 7.90 (d, 1H, = 8.2 Hz), 7.68 (dd, 1H, = 3.2 Hz & 9.4 Hz), 7.57 (br, 1H), 7.28-7.23 (m, 2H), 7.22-7.18 (m, 1H), 7.11-7.05 (m, 2H), 6.90 (d, 1H, = 7.6 Hz), 5.19-5.11 (m, 2H), 4.96-4.90 (m, 1H), 4.77-4.71 (m, 1H), 4.63- 4.58 (m, 1H), 4.39-4.32 (q, 2H, = 7.0 Hz), 4.27 (d, 1H, = 8.8 Hz), 3.95 (d, 1H, = 8.8 Hz), 2.88 (s, 3H), 2.47-2.36 (m, 2H), 1.39 (t, 3H, = 7.1 Hz); 13C NMR (75 MHz, CDC13): δ
179.4, 166.3, 159.7, 155.4, 150.9, 142.9, 139.9, 138.9, 136.6, 132.6, 131.7, 130.3, 129.8, 128.5, 124.3, 124.5, 123.2, 123.0, 121.9, 109.9, 78.8, 76.3, 66.9, 61.3, 54.7, 53.2, 41.1, 25.0, 14.2; MASS (ESI):m/z 580 [M+Na]+.
Example 10
Ethyl 2-methyl-6-(2-((l-(2-oxo-2 4^5 6'-tetrahydrospiro[indoline-3,3'-pyran]-5'-yl)-lH- l,2,3-triazol-4-yl)methoxy)-phenyl)nicotinate (AC-B)
1H NMR (300 MHz, CDC13): δ 8.11 (d, 1H, = 8.2 Hz), 8.06 (br, 5H), 7.84 (dd, 1H, = 1.6 Hz, 7.6 Hz), 7.79 (s, 1H), 7.77 (d, 1H, = 8.2 Hz), 7.73-7.70 (m, 0.5H), 7.55-7.52 (m, 0.5H), 7.42-7.38 (m, 1H), 7.19-7.11 (m, 3H), 6.89 (dt, 1H, = 0.9 Hz & 7.6 Hz), 6.84 (d, 1Η, = 7.7 Hz), 6.80 (d, 1H, = 7.3 Hz), 5.34 (dd, 2H, = 12.2 Hz & 17.0 Hz), 4.77-4.66 (m, 2H), 4.61 (dd, 1H, 7 = 5.1 Hz & 14.0 Hz), 4.34 (q, 2H, = 7.0 Hz), 4.05 (d, 1H, = 8.8 Hz), 3.89 (d, 1H, = 8.8 Hz), 2.88 (s, 3H), 2.53 (dd, 1H, = 6.4 Hz & 13.1 Hz), 1.91 (dd, 1H, = 9.6 Hz, 13.1 Hz), 1.36 (t, 3H, J = 7.0 Hz); 13C NMR (75 MHz, CDCI3): δ 180.3, 166.7, 159.4, 157.8,
155.8, 144.2, 140.0, 138.0, 132.1, 130.5, 128.9, 128.4, 124.2, 123.2, 122.5, 122.1, 121.8,113.1, 109.6, 78.2, 77.1, 76.8, 62.8, 61.0, 52.4, 40.4, 29.6, 25.0, 14.2; MASS (ESI): m/z 539 [M+Na]+.
Example 11
Ethyl 6-(5-chloro-2-((l-(2-oxo-2',4',5',6'-tetrahydrospiro [indoline-3,3'-pyran]-5'-yl)-lH- l,2,3-triazol-4-yl)methoxy)-phenyl)- 2-methyl nicotinate (AC-C)
1H NMR (300 MHz, CDCI3): δ 8.25 (d, 1H, = 8.2 Hz), 7.85 (d, 1H, = 8.2 Hz), 7.78 (dd, 1H, = 3.2 Hz & 9.4 Hz), 7.69 (br, 1H), 7.5 (s, 2H), 7.28-7.21 (m, 1H), 7.09-7.05 (m, 2H), 6.95-6.91 (d, 1H, J = 7.6 Hz), 4.95 (s, 2H), 4.73-4.68 (m, 1H), 4.62-4.57 (m, 1H), 4.43-4.39 (m, 1H), 4.26-4.24 (q, 2H, = 7.0 Hz), 3.95 (d, 1H, = 8.8 Hz), 2.88 (s, 3H), 2.37-2.36 (m, 2H), 1.39 (t, 3H, = 7.1 Hz); 13C NMR (75 MHz, CDC13): δ 179.4, 166.3, 159.7, 155.4,
150.9, 142.9, 138.9, 139.9, 136.0, 132.5, 130.7, 130.3, 129.8, 128.5, 124.5, 124.5, 123.2, 123.0, 121.9, 78.8, 76.3, 66.9, 61.3, 54.7, 53.2, 41.1, 25.0, 14.2; MASS (ESI): m/z 597
[M+Na]+.
Example 12
Ethyl 6-(3,5-dichloro-2-((l-(2-oxo-2',4',5',6'-tetrahydrospiro-[indoline-3,3'-pyran]-5'-yl)-lH- l,2,3-triazol-4-yl)methoxy)phenyl)-2-methyl nicotinate (AC-D)
1H NMR (300 MHz, CDC13) : δ 8.26 (d, 1H, = 8.0 Hz), 7.82 (d, 1H, = 8.0 Hz), 7.76 (dd, 1H, = 0.9 Hz & 2.8 Hz), 7.67 (s, 1H), 7.48 (dd, 1H, = 0.9 Hz & 2.5 Hz), 7.28-7.23 (m, 3H), 7.08 (t, 1H, = 7.4 Hz), 6.93 (d, 1H, = 8.5 Hz), 4.97 (s, 2H), 4.76-4.67 (m, 2H), 4.60- 4.55 (m, 1H), 4.39 (q, 2H, = 7.6 Hz), 4.25 (d, 1H, = 8.8 Hz), 3.96 (d, 1H, = 8.8 Hz), 2.90 (s, 3H), 2.38-2.26 (m, 2H), 1.41 (t, 3H, = 7.6 Hz); 13C NMR (75 MHz, CDC13): δ 178.5, 166.8, 159.5, 155.7, 151.8, 142.9, 139.7, 138.9, 136.2, 132.5, 130.7, 130.4, 129.8, 128.5, 124.5, 124.5, 123.3, 123.0, 121.9, 109.9, 78.8, 76.3, 66.9, 61.5, 54.7, 53.2, 41.1, 25.1, 14.1; MASS (ESI):m/z 631 [M+Na]+.
Example 13
2-((l-(2-oxo-2', 4', 5', 6'-tetra ydrospiro[indoline-3,3'-pyran]-5'-yl)-lH-l,2,3-triazol-4-yl) methoxy)benzaldehyde (AC-E)
1H NMR (300 MHz, CDC13): δ 10.49 (s, 1H), 7.92 (s, 1H), 7.85 (dd, 1H, 7 = 1.6 Hz & 7.6 Hz), 7.63 (br, 1H), 7.59-7.55 (m, 1H), 7.28-7.23 (m, 1H), 7.20 (d, 1H, = 8.3 Hz), 7.07 (q, 2H, J = 7.3 Hz), 6.91 (d, 1H, J = 7.6 Hz), 5.37 (s, 2H), 4.79-4.74 (m, 1H), 4.69-4.63 (m, 1H), 4.25 (d, 1H, J = 8.8 Hz), 3.96 (d, 1H, J = 8.8 Hz), 2.96 (s, 0.5H), 2.89 (s, 0.5H), 2.40-2.34 (m, 1H), 2.33-2.27 (m, 1H); 13C NMR (75 MHz, CDC13): δ 190.0, 182.3, 162.2, 142.1, 140.9, 139.8, 135.0, 131.2, 126.5, 125.5, 124.8, 123.1, 122.7, 122.6, 114.5, 110.2, 75.4, 73.2 71.0, 56.6, 51.1, 32.4; IR: 2959, 2870, 1720, 1691, 772 cm"1; MASS (ESI): m/z All [M+Na]+. Example 14
5'-(4-(Pyridin-2-yl)-lH-l,2,3-triazol-l-yl)-2',4',5',6'-tetrahydrospiro[indoline-3,3'-pyran]-2- one (AC-F)
1H NMR (300 MHz, CDC13): δ 10.18 (br, 1H), 8.59 (br, 1H), 8.41 (s, 1H), 8.13 (d, 1H, = 7.7 Hz), 7.80 (t, 1H, = 7.5 Hz), 7.33-7.15 (m, 3H), 7.0 (t, 1H, = 7.5 Hz), 6.91 (d, 1H, = 7.7 Hz), 4.90-4.70 (m, 3H), 4.23 (d, 1H, = 8.6 Hz), 3.94 (d, 1H, = 8.6 Hz), 2.42-2.30 (m, 2H); 13C NMR (75 MHz, CDC13): δ 178.2, 159.1, 148.7, 140.6, 136.2, 132.3, 127.6, 122.7, 122.1, 122.0, 122.0, 121.8, 119.4, 109.4, 78.0, 75.7, 54.2, 52.8, 40.4; IR: 3017, 2871, 1712, 1601, 1217, 771cm"1; MASS (ESI): m/z 370 [M+Na]+.
Example 15
5'-(4-(Hydroxy(phenyl)methyl)-lH-l,2,3-triazol-l-yl)-2 4^5 6'-tetrahydrospiro[indoline-3,3'- pyran]-2-one (AC-G)
1H NMR (300 MHz, CDC13) : δ 8.10 (d, 1H, = 7.1 Hz), 7.93 (br, 1H), 7.88-7.80 (m, 1H), 7.68-7.30 (m, 3H), 7.07 (t, 1H, J = 7.7 Hz), 6.90 (d, 1H, J = 6.7 Hz), 4.99-4.52 (m, 3H), 4.31-4.08 (m, 1H), 3.99-3.88 (m, 1H), 2.50-2.19 (m, 2H); 13C NMR (75 MHz, CDC13): δ 179.8, 139.9, 133.2, 130.5, 130.0, 128.9, 128.6, 128.6, 128.5, 128.3, 126.5, 123.2, 122.9, 110.1, 78.7, 76.3, 64.2, 54.7, 53.4, 29.6; IR: 3019, 1710, 1214, 745cm"1; MASS (ESI): m/z 399 [M+Na]+.
Example 16
5'-(4-((2-Nitrophenoxy)methyl)-lH-l,2,3-triazol-l-yl)-2',4',5',6'-tetrahydrospiro -[indoline- 3,3'-pyran]-2-one (AC-H) 1H NMR (300 MHz, CDCI3) : δ 8.18-8.14 (m, IH), 7.85-7.92 (m, IH), 7.61-7.55 (m, IH), 7.58-7.51 (m, IH), 7.17-7.05 (m, 2H), 6.94-6.90 (m, IH), 6.80-6.72 (m, 2H), 6.02-5.98 (m, IH), 4.80-4.67 (m, 3H), 4.27-4.21 (m, 3H), 4.18-4.10 (m, IH), 3.98-3.90 (m, IH), 3.51-3.47 (m, IH), 2.39-2.32 (m, 2H); 13C NMR (75 MHz, CDC13): δ 180.1, 155.4, 144.6, 143.3, 142.8, 140.2, 129.3, 127.8, 126.5, 123.1, 122.1, 114.5 109.5, 104.6, 99.6, 78.2, 75.7, 54.2, 53.4, 52.0, 35.4; MASS (ESI): m/z AAA [M+Na]+.
Example 17
5'-(4-((4-Chloro-2-methylphenoxy)methyl)- lH-l,2,3-triazol- l-yl)-2',4',5',6'-tetra
hydrospiro[indoline-3,3'-pyran]-2-one (AC-I)
1H NMR (300 MHz, CDCI3) : δ 8.80 (br, IH), 7.85 (s, IH), 7.77-7.56 (m, 2H), 7.36-7.19 (m, 2H), 7.13-6.85 (m, 3H), 5.19 (s, 2H), 4.82-4.60 (m, 3H), 4.22 (d, IH, = 8.6 Hz), 3.94 (d, IH, = 8.6 Hz), 2.47-2.40 (br, IH), 2.37-2.28 (m, IH), 2.19 (s, 3H); 13C NMR (75 MHz, CDCI3): δ 179.7, 154.8, 144.0, 132.4, 130.4, 129.7, 128.5, 128.2, 128.0, 126.3, 123.9, 123.1, 122.8, 112.6, 110.0, 78.7, 76.3, 62.2, 54.7, 53.3, 40.9, 16.1 ; MASS (ESI): m/z 447 [M+Na]+. Example 18
5'-(4-((3-Hydroxyphenylamino)methyl)- lH-l,2,3-triazol- l-yl)-2',4',5',6'- tetrahydrospiro[indoline-3,3'-pyran]-2-one (AC-J)
1H NMR (300 MHz, CDCI3) : δ 9.57 (br, IH), 8.13 (br, IH), 7.77-7.72 (m, IH), 7.36-7.31 (m, IH), 7.27-7.19 (m, 2H), 7.07-6.96 (m, 2H), 6.95-6.90 (m, IH), 6.27-6.19 (m, 2H), 4.79- 4.60 (m, 3H), 4.49-4.26 (m, 3H), 4.21-4.14 (m, IH), 3.94-3.88 (m, IH), 2.30-2.23 (m, IH), 2.22-2.16 (m, 2H); 13C NMR (75 MHz, CDCI3): δ 180.1, 158.5, 148.7, 146.5, 143.3, 129.3,
128.5, 126.8, 122.5, 122.1, 122.3, 109.5, 107.7, 106.6, 99.6, 77.2, 75.7, 55.2, 51.8, 42.3, 35.4; IR: 3476, 2922, 1733, 1232, 1018, 749cm"1; MASS (ESI): m/z A IA [M+Na]+.
Example 19
5'-(4-((3-Aminophenoxy)methyl)-lH-l,2,3-triazol- l-yl)-2^4^5 6'-tetra ydrospiro[indoline- 3,3'-pyran]-2-one (AC-K)
1H NMR (300 MHz, CDC13): δ 12.12-12.08 (m, IH), 8.08 (brs, IH), 7.98-7.92 (m, IH), 7.91- 7.85 (m, IH), 7.58-7.51 (m, IH), 7.17-7.05 (m, 2H), 6.95-6.90 (m, IH), 6.80-6.72 (m, 2H), 6.02-5.98 (m, IH), 4.80-4.65 (m, 3H), 4.27-4.20 (m, 3H), 4.18-4.10 (m, IH), 3.98-3.90 (m, IH), 3.51-3.47 (m, IH), 2.39-2.30 (m, 2H); 13C NMR (75 MHz, CDC13): δ 179.0, 157.4,
148.6, 146.3, 145.8, 140.2, 129.3, 127.8, 122.5, 122.1, 122.1, 109.5, 104.7, 104.6, 99.6, 78.2, 75.7, 54.2, 52.4, 52.3, 40.0; MASS (ESI): m/z A IA [M+Na]+.
Example 20
5'-(4-((5-Bromo-3-nitropyridin-2-yloxy)methyl)- lH- l,2,3-triazol- l-yl)-2',4',5',6'-tetrahydro spiro[indoline-3,3'-pyran]-2-one (AC-L)
1H NMR (300 MHz, CDC13): δ 8.41 (d, 1H, = 2.4 Hz), 8.27 (d, 1H, = 2.4 Hz), 8.15 (s, 1H), 7.29-7.20 (m, 3H), 7.06 (t, 1H, J = 7.5 Hz), 6.97-6.90 (m, 1H), 5.44-5.23 (m, 2H), 4.81- 4.64 (m, 3H), 4.23 (d, 1H, = 8.6 Hz), 3.95(d, 1H, = 8.8 Hz), 2.41-2.22 (m, 2H); 13C NMR (75 MHz, CDCI3): δ 180.1, 155.8, 154.7, 140.3, 138.7, 135.8, 130.6, 125.2, 124.1, 122.0, 121.8, 110.2, 101.9, 74.4, 73.5, 71.5, 56.2 51.8, 30.8; MASS (ESI): m/z 524 [M+Na]+.
Example 21
Ethyl 2-methyl-6-(2-((l-((2'-oxo-4,5-dihydro-2H-spiro[furan-3,3'-indoline]-5-yl)methyl)- lH- l,2,3-triazol-4-yl)methoxy)phenyl)nicotinate (AB-O)
Yield=72%; Semi solid; IR (neat): vmax 3206, 2929, 1716, 1584, 1265, 1010, 771 cm"1; 1H NMR (300 MHz, CDCI3): δ 8.52 (s, 1H), 8.25 (dd, J = 8.1Hz, 3.7Hz, 1H, ), 7.84-7.93 (m, 3H), 7.46-7.3 l(m, 4H), 6.80-6.86 (m, 2H), 5.39 (d, 7 = 9.0Hz ,4.7Hz, 1H), 4.57-4.63 (m, 3H), 4.31-4.37 (m, 2H), 4.16 (dd, J = 7.1Hz, 6.7Hz,lH), 3.91 (dd, J = 8.8Hz, 3.7Hz, 1H), 2.87 (d, = 4.3 Hz, 3H), 2.51 (d, = 4.3 Hz, 3H), 2.51-2.34 (m, 1H), 1.34- 1.41 (m, 3H); 13CNMR (75 MHz, CDCI3): δ 180.0, 166.6, 159.3, 157.7, 144.2, 140.2, 138.0, 132.5, 132.0, 131.4, 130.5, 128.6, 128.7, 128.4, 128.0, 123.9, 122.8, 122.3, 113.0, 110.2, 109.8, 78.7, 78.1, 62.4, 61.1, 64.4, 52.5, 40.3, 25.0, 14.1 ; MS (ESI): m/z 540 [M+H]+; HRMS [M+H]+: calcd for C30H30O5N5 540.2202, found 540.2241.
Example 22
Ethyl 6-(5-fluoro-2-((l-((2'-oxo-4,5-dihydro-2H-spiro[furan-3,3'-indoline]-5-yl)methyl)- lH- 1 ,2,3-triazol-4-yl)methoxy)phenyl)-2-methylnicotinate (AB-M)
Yield = 77%; Semi solid; IR (neat): vmax 3215, 2930, 1717, 1472, 1261, 771, 680 cm"1; 1H NMR (300 MHz, CDC13) : δ 8.31 (s, 1H), 7.79-7.85 (m, 3H), 7.25-7.02 (m, 5H), 6.91- 6.84 (m, 2H), 5.42-5.49 (m, 2H), 4.81-4.89 (m, 3H), 4.52-3.92 (m, 4H),2.87 (s, 3H), 2.54 (dd, J = 6.5 Hz, 4.7Hz, 1H), 2.34 (dd, = 7.6 Hz, 3.7Hz, 1H), 1.37 (t, J = 1.1 Hz, 3H); 13CNMR (75 MHz, CDCI3): δ 180.8, 166.5, 159.4, 156.2, 152.0, 143.4, 140.3, 138.3, 132.0, 130.1, 129.1, 128.1, 124.5, 123.3, 122.9, 122.0, 117.0, 116.1, 114.4, 109.1, 78.5, 78.1, 63.3, 61.0, 54.4, 52.4, 40.1, 29.1, 24.1, 13.1 ; ESI-MS: m/z 558[M+H]+; HRMS[M+H] +: calcd for CsoHssOsNsFNa 580.1932, found 580.1932.
Example 23
Methyl-6-(5-chloro-2-((l-((2'-oxo-4,5-dihydro-2H-spiro[furan-3,3'-indoline]-5-yl)methyl)- 1 H- 1 ,2,3 -triazol-4-yl)methoxy)phenyl)-2-methylnicotinate ( AB -N)
Yield = 71%; Solid; mp: 120- 122 °C; IR (neat): vmax 3218, 2926, 1718, 1619, 1557, 1263, 771, 678 cm"1; 1H NMR (300 MHz, CDC13): δ 8.13 (d, = 8.2 Hz, 1H), 7.83-7.63(m, 1H), 7.25- 7.02 (m, 4H), 6.91 (dd, J = 8.0 Hz, 3.7Hz, 1H), 6.81-6.89 (m, 1H), 5.29 (d, J = 8.0 Hz, 2H), 4.78-4.54 (m, 3H), 4.32-4.38 (m, 2H), 4.12-3.92(m, 3H), 2.87 (s, 3H), 2.52-2.34(m,2H), 1.37 (s, 3H); 13CNMR (75 MHz, CDC13): δ 180.8, 166.5, 159.4, 156.2, 152.0, 143.4, 140.3, 138.3, 132.0, 130.1, 129.4, 128.1, 124.5, 122.9, 122.0, 117.0, 116.1, 114.4, 109.1, 123.3, 78.5, 78.1, 63.3, 61.0, 54.4, 52.4, 40.1, 29.1, 13.2; ESI-MS: m/z 560 [M+H]+; HRMS: calcd for C29H27O5N5CI 560.1657, found 560.1692.
Example 24
2-((l-((2'-Oxo-4,5-di ydro-2H-spiro[furan-3,3'-indoline]-5-yl)methyl)-lH-l,2,3-triazol-4- yl)methoxy)benzaldehyde (AB-P)
Yield = 71%; Semi solid; IR (neat): vmax 2930, 2868, 1714, 1689, 1599, 1471, 1236, 1005, 754, 680 cm"1; 1H NMR (300 MHz, CDCI3): δ 10.52 (s, 1H), 8.53 (s, 1H), 7.91 (s, 1H), 7.86 (dd, J = 7.5 Hz, 6.7Hz, 2H), Ί .55-1.62 (m 1H), 7.30-6.85 (m, 3H), 5.40 (s, 2H), 4.84-4.62 (m, 4H), 4.17-3.96 (m, 3H), 2.58 (dd, = 6.8 Hz, 2.7Hz, 1H), 1.95 (dd, = 6.1 Hz, 4.7Hz, 1H); 13CNMR (75 MHz, CDCI3): δ 189.6, 189.5, 160.5, 143.4, 136.0, 132.0, 136.0, 128.5, 128.7, 124.6, 123.1, 122.4, 121.3, 113.0, 110.0, 78.1, 76.6, 62.4, 54.3, 52.8, 40.6, 29.7; ESIMS: m/z All [M+Na]+; HRMS: calcd for C22H20O4N4Na 427.1349, found 427.1376.
Example 25
Ethyl6-(3,5-dichloro-2-((l-((2'-oxo-4,5-dihydro-2H-spiro[furan-3,3'-indoline]-5-yl)methyl)- 1 H- 1 ,2,3 -triazol-4-yl)methoxy)phenyl)-2-methylnicotinate ( AB - W)
Yield = 78%; Solid; mp: 132- 135 °C; IR (neat): vmax 3197, 2925, 1719, 1712, 1620, 1468, 1078, 965, 754, 680 cm"VH NMR (300 MHz, CDC13):5 8.81 (s, 1H), 8.23 (d, = 8.1 Hz, 1H), 7.81 (d, = 8.1 Hz, 1H), 7.77 (dd, J= 5.65 Hz, 2.6 Hz, 2H), 7.44-7.49 (m, 1H), 7.26- 6.86 (m, 5H), 5.0 (d, = 7.5 Hz, 2H), 4.76-4.58 (m, 2H), 4.40-4.42 (m, 2H), 4.18-4.25 (m, 2H), 3.85-3.91 (m, 1H), 2.89 (d, = 3.02 Hz, 2H), 2.52-2.58 (m 1H), 2.28-2.33 (m,lH), 1.37 (s, 3H); 13CNMR (75 MHz, CDCI3): δ 180.8, 179.7, 166.3, 159.6, 155.4, 151.0, 143.1, 140.3, 138.9, 136.4, 132.5, 130.7, 130.3, 129.7, 128.4, 124.7, 123.0, 122.5, 121.8, 109.8, 78.8, 78.26, 67.2, 61.2, 60.3, 54.5, 52.4, 40.5, 24.9, 14.2; ESIMS: m/z 608 [M+H]+; HRMS: calcd for C3oH2705N5Cl2Na 630.1249, found 630.1281.
Example 26
Ethyl-l-((2'-oxo-4,5-di ydro-2H-spiro[furan-3,3'-indoline]-5-yl)methyl)-lH-l,2,3-triazole-4- carboxylate (AB-F)
Yield = 88%; White solid; mp: 133-134 °C; IR (neat): vmax 3138, 2926, 2855, 1737, 1685, 1607, 1250, 1041,7 54 cm"1; 1H NMR (300 MHz, CDC13): δ 8.40 (s, IH), 7.96 (dd, = 4.5 Hz, 5.2 Hz, 2H), 7.40-7.18 (m, 3H), 7.0-7.16 (m, 2H), 4.90-4.63 (m, 3H), 4.43-4.49 (m , 2H), 4.18-4.25 (m, IH), 3.94-4.01 (m, IH), 2.57-2.62 (m, IH), 1.97-2.04 (m,lH), 1.31-1.36 (m, 3H); 13CNMR (75 MHz, CDC13): δ 178.1, 167.4, 133.2, 129.0, 128.7, 125.4, 122.9, 123.3, 110.2, 107.1, 78.7, 78.0, 61.4, 60.6, 52.0, 41.4, 14.3; ESIMS: m/z 365 [M+Na]+; HRMS: calcd for Ci7H20N4O4Na 365.1632, found 365.1642.
Example 27
5-((4-Phenyl-lH-l,2,3-triazol-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]-2'-one (AB-C)
Yield = 78%; White solid; mp: 175-177°C; IR (neat): vmax 3229, 2925, 2855, 1713, 1620, 1470, 1221, 1077, 769, 694 cm"1; 1H NMR (300 MHz, DMSO):5 9.22 (s, IH), 7.28 (d, = 7.1 Hz, IH), 7.02 (d, = 7.17 Hz, 2H), 7.01-7.09 (m, 3H), 6.29 (dd, = 7.3 Hz, IH), 6.02-6.09 (m, 3H), 3.94-4.01 (m, 3H), 3.22 (dd, = 8.7 Hz,3.8Hz, IH) 3.12 (dd, = 8.8 Hz,1.9Hz, IH), 1.72 (dd, = 6.2 Hz,1.9Hz, IH), 1.18 (dd, = 8.12 Hz,3.9Hz IH); 13CNMR (300 MHz, DMSO): δ 179.4, 146.4, 140.6, 131.2, 129.7, 128.1, 127.8, 127.3, 127.2, 127.1, 127.6, 120.4, 120.0, 108.9, 77.4, 75.8, 66.2, 53.4, 51.8, 39.7; ESI-MS: m/z 347 [M+H]+; HRMS: calcd for C20H18O2N4Na 369.1325, found 369.1325.
Example 28
5-((4-(Hydroxymethyl)-lH-l,2,3-triazol-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'- indolin]-2'-one (AB-G)
Yield = 78%; Yellow liquid; IR (neat): vmax 3352, 2956, 2922, 2852, 1712, 1468, 1220, 772 cm"1; 1H NMR (300 MHz, CDCI3): δ 9.94 (s, IH), 7.84 (d, = 8.1 Hz, IH), 7.20-6.83 (m, 4H), 4.80-4.58 (m, 5H), 3.93-3.98 (m, 2H), 3.08 (br, s, IH), 2.47-2.53 (m, IH), 2.20-2.28 (m,lH); 13CNMR (75 MHz, CDC13): δ 181.2, 141.6, 131.9, 127.4, 124.0, 123.4, 121.9, 111.1, 78.2, 75.5, 68.3, 54.6, 52.2, 40.4, 36.5; ESI-MS: m/z 323 [M+Na]+; HRMS: calcd for Ci5Hi603N4Na 323.1114, found 323.1114.
Example 29
5-((4-(2-Hydroxypropan-2-yl)-lH-l,2,3-triazol-l-yl)methyl)-4,5-dihydro-2H-spiro[furan- 3,3'-indolin]-2'-one (AB-E)
Yield = 78%; Thick liquid; IR (neat): vmax 3272, 2972, 2927, 1711, 1620, 1221, 760,680 cm" VHNMR (300 MHz, CDCI3): δ 10.06 (s, IH), 7.76 (d, = 13.2 Hz, IH), 7.48 (s, IH), 7.17- 7.24 (m, IH), 6.91-6.89 (m, 2H), 4.80-4.60 (m, 3H), 4.18-3.90 (m, 2H), 2.53 (dd, J = 6.7 Hz, 4.2Hz, 2H), 2.22-2.28 (m, IH), 1.62 (d, = 13.5 Hz, 6H); 13CNMR (75 MHz, CDC13): δ 182.2, 155.9, 140.6, 130.9, 128.4,125.1, 123.0, 122.4, 110.1, 77.5, 76.3, 68.3, 54.6, 52.2, 40.4, 30.5, 30.4; ESI-MS: m/z 351 [M+Na]+; HRMS: calcd for Ci7H2i03N4 329.1604, found
329.1608.
Example 30
5-((4-(4-Nitrophenyl)-lH-l,2,3-triazol-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'- indolin]-2'-one (AB-D)
Yield = 70%; Thick liquid; IR (neat): vmax 3272, 2926, 2855, 1715,1655, 1517, 1338, 854, 680 cm"1; 1HNMR (300 MHz, CDC13): δ 8.62( s, 1H), 8.51-8.58(m, 5H), 7.34-7.41(m, 2H), 7.22-7.28(m, 2H), 4.79-4.84(m, 2H), 4.50-4.56(m, 3H), 2.47-2.52(m, 2H); 13CNMR (75 MHz, CDC13): δ 181.2, 162.6, 160.8, 140.3, 138.1, 134.5, 129.2, 129.2,122.8, 115.3, 115.1, 77.9, 77.2, 74.3, 54.3, 53.8, 52.2, 51.8, 51.4, 42.4;ESJ S: m/z 392 [M+H]+; HRMS: calcd for C2oHi704N5Na 414.175, found 414.1759.
General procedure for spirooxindole-amines (8)
To the spiro-iodo compound 6a (0.5 mmol) in Dry DMF was added secondary amine (0.5 mmol) and dry K2C03 (0.6 mmol), under inert conditions, at room temperature and stirred for 15 min. Then the reaction mixture was heated to 80 °C and maintained with vigorous stirring for about 12 h, after completion of the reaction (monitored by TLC), solvent was evaporated in vacuum and the residue was purified by column chromatography with hexane/EtOAc (3:7) to elute product 8.
Example 31
5-((4-Phenylpiperazin-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]-2'-one (AB-X) Yield = 78%; Solid; mp: 130- 132 °C; IR (neat): vmax 3738, 2924, 2824, 1713, 1599, 1467, 1233, 1011, 755, 621 cm"1; 1H NMR (300 MHz, CDC13) : δ 8.57 (s, 1H), 7.27-7.34 (m, 5H), 7.04-7.14(m, 4H), 4.25-3.94 (m, 4H), 3.22-3.28 (m, 3H), 2.78-2.85 (m, 4H), 2.58-2.68 (m, 2H), 2.11-2.18 (m, 2H); 13CNMR (75 MHz, CDC13): δ 180.3, 160.7, 151.8, 140.2, 130.7, 129.0, 128.1, 123.0, 122.9, 119.7, 117.0, 116.0, 109.9, 78.4, 70.2, 62.3, 62.3, 54.7, 53.7, 48.9, 42.9, 40.7; ESIMS: m/z 364 [M+H]+; HRMS: calcd for C22H2602N3 364.2016, found 364.2019.
Example 32
5-(Morpholinomethyl)-4, 5-dihydro-2H-spiro [furan-3, 3'-indolin]-2'-one (AB-K)
Yield = 83%; white solid; mp: 126- 128 °C; IR (neat): vmax 3225, 2925, 1715, 1470, 1219, 772, 626 cm"1; 1H NMR (300 MHz, CDC13) :δ 8.2 (s,lH), 7.32 (d, = 7.5 Hz, 1H), 7.24 (t, = 7.3 Hz, 2H), 7.1 (d, = 7.9 Hz, 1H), 4.49-4.54 (m, 1H), 4.82 (dd, = 5.2 Hz,1.2Hz, 2H), 3.75- 3.82 (m, 4H), 2.97 (s, 1H), 2.88 (s, 1H), 2.78 (dd, = 12.1 Hz, 3.1 Hz, 1H), 2.60-2.67 (m, 4H), 2.59 (dd, = 6.0 Hz,4.3Hz, 1H); 13CNMR (75 MHz, CDC13): δ 180.3, 140.1, 133.1, 128.0, 124.1, 122.7, 122.5, 109.9, 76.1, 77.4, 78.0, 66.6, 62.1, 54.0, 53.4, 42.6; ESIMS: m/z 289 [M+H]+; HRMS: calcd for Ci6H20O3N2Na 311.1363, found 311.1366.
Example 33
5-((4-(Pyridin-2-yl)piperazin-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]-2'-one (AB-S)
Yield = 78%; viscous liquid; IR (neat): vmax 2922, 2856, 1712, 1467, 1220, 1121, 1034, 772 cm"1; 1H NMR (300 MHz, CDC13): δ 8.86 (s,lH), 8.20 (dd, = 8.7Hz, 1.5 Hz, 2H), 7.28-7.21 (m, 3H), 7.06-6.92 (m, 3H) 4.54 (m, 1H), 4.09 (s, 2H), 3.68-3.42 (m, 5H), 2.98-2.60 (m, 6H), 2.0 (dd, = 9.2 Hz, 3.2 Hz, 1H); 13CNMR (75 MHz, CDC13): δ 181.3, 171.4, 159.3, 147.7, 137.4, 134.2, 133.1, 127.9, 122.8, 122.2, 113.2, 109.8, 107.0, 78.0, 76.2, 62.1, 60.3, 54.5, 53.5, 49.9, 43.1; ESIMS: m/z 365 [M+H]+; HRMS: calcd for C2iH2502N4 365.1957 found 365.1972.
Example 34
5-((4-(2-Methoxyphenyl)piperazin-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]-2'- one (AB-U)
Yield = 78%; solid; 143-145 °C; IR (neat): vmax 3246, 2946, 2123, 1714, 1472, 1245, 1025, 765, 682 cm" YHNMR (300 MHz, CDC13): δ 8.48 (s, 1H), 7.42-7.26 (m, 5H), 6.98-7.10 (m, 3H), 4.51-4.59 (m, lH),4.16(s,2H), 3.87 (s,3H), 3.20-3.10 (m,4H), 2.90 (ddJ = 8.9 Hz, 3.9Hz,lH), 2.78-2.84 (m,4H), 2.68 (dd, =3.9Hz,3.1 Ηζ,ΙΗ), 2.59 (dd, = 6.0 Hz, 3.1 Ηζ,ΙΗ), 1.92 (dd, = 3.5 Hz, 3.1 Hz,lH):13C NMR (75 MHz, CDC13):(5179.1, 152.2,144.8,141.3, 142.0,134.0,128.3, 129.1, 126.5,122.9,122.6,120.9,118.1,10 8.4,74.1,77.1,62.4,60.35, 55.29, 53.9, 53.6, 50.47, 43.7; ESI-MS: m/z 394 [M+H]+; HRMS : calcd for C23H2803N3 394.2127 found 394.2125.
Example 35
5-((4-(4-Methoxyphenyl)piperazin-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]-2'- oneB (AB-T)
Yield = 78%; Solid; mp: 146-147 °C; IR (neat): vmax 3234, 2925, 2100, 1713, 1470, 1240, 751, 625 cm"VH NMR (300 MHz, CDC13): δ 8.46 (s, 1H), 7.34 (d, = 10.2 Hz, 3H), 7.24-7.08 (m, 1H), 6.87-6.93 (m, 6H), 4.51-4.58 (m, = 8.5 Hz, 1H), 4.10 (s, 2H), 3.78 (s, 3H), 3.19 (dd, = 3.9 Hz, 4H), 2.86-2.70 (m, 5H), 1.9 (dd, = 3.5 Hz, 1H); 13CNMR (300 MHz, CDC13): δ 192.1, 172.9, 155.9, 145.6, 140.0, 134.5, 128.1,127.9,123.9, 123.1, 122.9, 118.2, 114.4, 109.7, 78.1, 77.4, 62.1,62.0, 55.5, 53.8, 50.4, 43.4, 29.6; ESI-MS: m/z 393[M+H]+; HRMS: calcd for C23H2803N3 394.2105 found 394.2125.
Example 36 5-((4-Benzoylpiperazin-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]-2'-one (AB-Q) Yield = 78%; Thick liquid; IR (neat): vmax 3223, 2924, 2855, 1716, 1620, 1282, 1011, 753 cm"1; 1H NMR (300 MHz, CDC13): δ 8.61 (s, 1H), 7.41-7.49 (m, 4H), 7.29-7.35 (m, 3H), 7.60- 7.51 (m, 2H), 4.51-4.58 (m, 1H), 4.01-4.08 (m, 3H),3.87-3.93(m, 2H) 3.77-3.84 (m, 2H), 2.59-2.65 (m, 5H), 2.15 (d, = 5.2 Hz, 1H), 1.90 (dd, = 9.8 Hz, 2.3Hz, 1H); 13CNMR (75 MHz, CDC13): δ 181.0, 173.2, 140.2, 140.0, 135.9, 134.3, 129.6, 128.4, 128.0, 126.9,123.18, 122.7, 109.8, 78.0, 76.2, 66.7, 62.1, 55.5, 54.4, 53.8, 50.4, 43.1, 29.6; ESI-MS: m/z 392 [M+H]+; HRMS: calcd for C2 H2603N3 392.1950 found 392.1968.
Example 37
5-(Thiomorpholinomethyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]-2'-one (AB-L)
Yield = 78%; Thick liquid; IR (neat): vmax 2925, 1720, 1468, 1219, 772, 681 cm"1; 1HNMR (300 MHz, CDC13): δ 7.71 (br, s, 1H), 7.54 (dd, = 3.3 Hz, 2.1 Hz, 1H), 7.47-7.22 (m, 2H), 7.05 (d, = 7.6 Hz, 1H), 4.43-4.51 (m, 1H), 4.22 (dd, = 2.7 Hz, 1.2 Hz, 2H), 4.06 (d, = 3.2 Hz,1.3 Hz, 2H), 2.84-2.89 (m, 4H), 2.71-2.77 (m, 4H), 2.65 (dd, = 4.7 Hz, 3.3 Hz, 1H), 2.55 (dd, = 6.5 Hz, 2.1 Hz, 1H); 13CNMR (300 MHz, CDC13): δ 181.2, 140.1, 134.3, 130.9, 128.1, 123.1, 122.8, 109.8, 78.0, 77.3, 62.8, 61.1, 55.4, 43.3, 29.6, 27.7; ESI-MS: m/z 305 [M+H]+; HRMS: calcd for Ci6H20O2N2SNa 327.1136 found 327.1136.
Example 38
5-((4-Benzylpiperazin-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]-2'-one (AB-R) Yield = 78%; Solid; mp 141-143 °C; IR (neat): vmax 3215, 2926, 2812,1718,1684,1293,1011,745 cm"1; 1HNMR (300 MHz, CDC13): δ 7.87 (br, s, 1H), 7.24- 7.31 (m, 6H), 7.05-6.91(m, 2H), 4.38-4.42 (m, 1H), 4.21-4.02 (m, 2H), 3.53 (s, 2H), 3.35- 3.42 (m, 1H), 2.80-2.39 (m, 6H), 1.82-1.56 (m, 2H), 1.26-1.11 (m, 4H); 13CNMR (300 MHz, CDC13): δ 180.4, 139.9,133.1, 132.3, 132.1, 131.9, 128.4, 128.4, 123.1, 123.0, 122.1, 121.9, 109.9, 109.7, 80.7, 76.1, 69.5, 64.1, 62.5, 60.3, 54.9, 41.2, 38.0; ESIMS: m/z 378 [M+H]+; HRMS: calcd for C23H27N302Na 401.1266 found 401.1236.
Example 39
5-((4-(Bis(4-fluorophenyl)methyl)piperazin-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'- indolin]-2'-one (AB-V)
Yield = 78%; Thick liquid; IR (neat): vmax 3220, 2929, 1712, 1504, 1221, 1009, 771 cm"1; 1H NMR (300 MHz, CDC13): δ 8.43 (br, s, 1H), 7.40-7.20 (m, 6H), 7.06-6.86 (m, 6H), 4.46-4.53 (m, 1H), 4.23 (s, 1H), 4.04 (s, 2H), 2.80 (dd, = 8.3 Hz, 3.2Hz, 5H), 2.68-2.39 (m, 6H), 1.91 (dd, = 9.0 Hz, 7.1 Hz, 1H); 13CNMR (75 MHz, CDC13): δ 181.3, 162.6, 162.3, 160.7, 140.2, 140.1, 138.1, 138.1, 134.5, 129.2, 129.1, 128.0, 127.9,124.3,123.9, 122.1,115.3,115.1,109.7, 77.9,77.2, 74.3, 62.2,60.3, 54.5,53.8, 52.1, 51.4, 43.2; ESI-MS: m/z 490 [M+H]+; HRMS: calcd for CigHigOiNsFiNa 512.2098 found 512.1912.
The present invention provides new class of spirooxindoles which were synthesized from oxindole.
A program was initiated in the laboratory for the design and synthesis of novel spirooxindoles, which can serve as new chemical entities for drug discovery process. In these efforts new spirooxindoles have been synthesized and evaluated for GSK3P activity and the anticancer potential. The synthesis of these compounds has been carried out as described in Scheme 1 using oxindole as the starting substrate.
Biological activity
In vitro kinase assays screening of the small fragments archived in small molecule compounds library against GSK3P kinase activity has identified three potential fragments with IC50 values below 10 μΜ. The library of spiroxyindoles prepared based on the identified fragments in the present study was evaluated for their anti-kinase activity of GSK3p. The result indicates that ten compounds have potential to inhibit enzyme activity with Κ½ο<5μΜ. Though the compounds do not inhibit enzyme activity equivalent to commercially available inhibitors, observed IC50 values are better than LiCl standard GSK3P inhibitor. Glycogen Synthase Kinase 3β (GSK3P) is a serine/threonine kinase. It is a multifunctional enzyme, regulating several proteins involved in various signaling pathways including transcription factors, cell cycle/survival regulators and proto-oncogenes. Recent studies suggest that GSK3P may promote tumorigenesis in many cancer types. To confirm specificity of molecules inhibiting intracellular GSK3P, we have determined phosphorylation of glycogen synthase and accumulation of β-catenin. In the HEK293T cells treated with test compounds pGS levels decreased and increased levels of β-catenin confirms its stabilization due to decreased rate of proteasomal degradation. Pancreatic cancer cells have already been reported to express high active pool of GSK3p. Recent studies have revealed that the pancreatic cancer cells are resistant to TRAIL-induced apoptosis. The cytotoxicity results indicate that the compounds significantly increased sensitivity of PANC-1 cells to TRAIL. Further to confirm of cancer cells death by GSK3P inhibitors in combination with TRAIL, we have analyzed cell cycle progression of proliferating PANC-1 cells treated with different concentrations of compounds in the presence or absence of TRAIL. These results are in agreement with the previously reported results that the GSK3P inhibitors sensitize cancer cells to TRAIL induced cell death.
Biological screening table
Figure imgf000027_0001
Biological Activity and Screens
Molecular modelling:
The docked pose of potent GSK3b with available ligands C, O, and U was predicted by Autodock tools (ADT) and Autodock 4.2. The ligand coordinate file was generated using Chemdraw followed by minimization protocol, performed using steepest descent algorithm. Existing co-crystal structure with defined binding site of GSK3P with PDB ID 4AFJ having 1.98A resolution was retrieved from Protein Data Bank (PDB). Crystal structure was used for docking by removing the heteroatoms, hydrogen atoms and kollman charges were added to protein crystal structure using ADT. The pdb files of the ligands and protein structure were converted to pdbqt file using ADT. Docking studies were concentrated on the specific binding site known as ATP binding site. The docking results were further analyzed by using ADT and 3D visualization tool PyMOL. Activity of recombinant GSK3P is measured using ADP-Glo Kinase assay according to supplier's standard protocol with minor modifications in 384 well format. Briefly, kinase reaction mixture was prepared by adding 5 mg of recombinant enzyme, 2 μΜ ATP, 1 μg of enzyme substrate along with test compound or DMSO to a final volume of 5 μΐ. The kinase reaction was performed by incubating the mixture at room temperature for 17 mins. To correct reaction back ground a substrate blank reaction was also performed. ADP glow reagent (equal volume to reaction mixture) was added to the reaction mixture to deplete any unused ATP added to the kinase reaction and incubated at further 40 min at 27 °C. Then 10 μΐ of kinase detection reagent was added followed by incubation for 30 min at 27 °C to convert ADP to ATP and introduce luciferase to measure kinase activity. The luminescence was measured using luminometer (Perkin Elmer) which is directly proportional to ADP generated in kinase reaction. To determine the amount of ADP produced in kinase reaction and calculate specific activity of the enzyme, a conversion curve is prepared by serial dilution of ADP and ATP mixture. Specific activity of the GSK3P was calculated using the following equation.
(ADP (kinase reaction) - ADP (substrate blank)) in nmol) /
(Reaction time in min) x (GSK3P in mg).
Cell culture:
The cell pLen,(JZ)-Top.Luc, pLen-TopFlash.dGFP 293T stable cells were generated in myoclinic USA, panc-1 used in the current study were obtained from ATCC (Manassas, USA) and cultivated in DMEM (Sigma) supplemented with 10% fetal bovine serum, 100 units/mL penicillin and streptomycin. To avoid for mycoplasma contamination in cell cultures, cells were regularly tested using the MycoAlert Kit (Cambrex Bio Science Rock- land, Inc., Rockland, ME, USA).
Luciferase assay:
To determine inhibitory potential of hit molecules from primary screening against intracellular GSK3P, a reporter assay system has been established using pLenti(JZ)-Top.luc vector and HEK293T cells. The stable cell lines having integrated TCF/LEF luciferase reporter gene were selected in the presence of suitable antibiotics in the culture medium. To test activity of hits against GSK3P, stable cells were treated with different concentrations (0- 100 μΜ) of either compounds or standard positive controls. Cells were allowed to grow for further 24 hrs under standard culture conditions. Upon termination, cells were harvested directly in lx CC1 buffer. To determine luciferase activity, 5 μΐ of cell lysate was mixed with 20 μΙ_, of assay reagent (company name) containing luciferase substrate in a MTP well pate (384 well format) and incubated for 5 mins in dark. The luciferase activity was determined by measuring luminescence at wavelengths in multimode reader. The protein concentration of the lysates was estimated by Bradford assay to normalize luciferase activity with protein present in the lysates. Then the relative luciferase activity was calculated according to control cells treated with DMSO set to 1. All experiments were performed in triplicates and the mean values with + SD are presented here.
Cell viability:
Cell viability was measured using Sulforhodamine B assay. The cytotoxic effect of the compounds with GSK3P inhibitory potential was tested against PANC-1 cells. Initially, the PANC-1 cells were seeded in flat bottom 96-well plate (5000 cells/100 μί) in complete medium and cultured for 18 hrs under standard conditions. After 18 h the cells were treated with either the different concentration of compounds, standard drugs or DMSO as solvent control. The final DMSO concentration to cells was maintained < 2% in all cytotoxic assays. Each compound was tested in triplicate and the cytotoxicity was determined as the average of that triplicate. Further, the plates were incubated for another 48 h in an incubator maintained at 37 °C with a constant supply of 5% C02. The cells were fixed using 10% TCA and incubated for 1 h at 4 °C. Then the plates were rinsed carefully with MQ water and air dried at room temperature. To all the wells of MTP plate 0.057% SRB solution was added and kept for approximately 30 min before it was rinsed off using 1% acetic acid. The plates were then air dried and 100 μΐ^ of 10 mM tris base was added to each well to solubilize the SRB before absorbance was measured at 510 nm. The measure of absorbance is directly proportional to cell growth and is thus used to calculate % reduction in the rate of cell proliferation according to control DMSO treated cells. All experiments were performed in triplicates; mean % age proliferation values with + SD were plotted here.
Treatment of PANC-1 cells with GSK3P inhibitors
The compounds inhibiting GSK3P activity in-vitro added to PANC-1 cells to determine their potential against intracellular GSK3P signalling pathways in cancer cells. Briefly, PANC-1 cells were plated in 60 mm plates (100,000 cells/ 4 mL) and cultured in the standard DMEM medium containing 10% serum. After culturing for 18 hrs, different concentrations of test compounds or standard controls prepared in DMSO were added to culture media to achieve final concentration 0 to 100 μΜ of compound to cells. Cells were further continued to grow for 48 h with constant supply of 5% C02 in humid incubator. The time course (0 to 48 h) experiment was carried out simultaneously with fixed concentration of compound showing 50% growth inhibition. At the end of the experiments, all cells were scraped directly in cold PBS and washed for 3 times before lysis directly in 2D lysis buffer (8M urea, 2M thiourea, 4% CHAPS, 4mM tris base and 65 mM DTT) for isolation of proteins to perform western blots. The protein concentration was determined by modified Bradford assay.
Western blotting:
To perform western blot, protein extracts were separated on 12% SDS-PAGE and electrophoretically (1 mA/cm membrane for 1 hr) transferred onto the nitrocellulose membrane by semi-dry method. After protein transfer, membrane was stained with Ponceau S solution (0.2% Ponceau S in 3% Trichloroacetic acid) for 2 min to see efficiency of electrophoretic transfer. The membrane was destained in ddH20 to visualize the protein bands and further destained with TBST (20 mM Tris, 138 mM NaCl, pH 7.6 and 0.1% Tween 20) for blotting. To avoid non-specific detection of target proteins, blotted membranes were blocked with BSA (3% BSA in TBST solution). All antibodies against specific protein targets of interest were purchased from Cell Signaling Technology, USA if not mentioned specifically. Then the blots were incubated with antibodies prepared in blocking buffer (1: 1000 Antibody to blocking buffer dilution) for 18 h with constant shaking at 4 °C. Three washing steps in PBS each for 5 mins removed the excess and non-specific antibodies. For visualization, blots were incubated with secondary antibody conjugated to either horseradish peroxidase or alkaline phosphatase [anti-(mouse IgG) or anti-(rabbit IgG), diluted 1:5000 in TBST] for 1 h at room temperature. After three washes in PBS, the reaction was developed either by addition of LumiGLO substrate (Cell Signaling Technology) followed by capturing on X-ray film or addition of chromogenic NBT (nitro-blue tetrazolium chloride) and BCIP (5-bromo-4-chloro-3'-indolyphosphate p-toluidine salt) substrate for alkaline phosphatase developing purple colored bands. The blots were scanned for densitometry performed using Image-J program.
NFkB Lucif erase assay:
In order to evaluate the effect of GSK3P inhibition on NF-kB activity, a reporter assay has been performed in the presence of identified inhibitors. To perform reporter assays, PANC-1 cells were cultured in a 48 well plate (16000 cells/well) overnight. Following day, cells were transfected with appropriate amount NF-kB reporter construct using lipofectamine 2000 as per manufacturer's protocol. After 16 hrs of post transfection cells were treated with identified GSK3P inhibitors and positive controls. After 48 h of treatment, cells were harvested in directly lx ccl buffer. For measuring lucif erase activity, 5 μΐ of cell lysate was mixed with 20 μΐ of assay reagent containing substrate for luciferase in MTP well plate as per manufacturer's protocol. The luciferase activity was determined by measuring luminescence at wavelengths in multimode reader. The protein concentration of the lysates was estimated by Bradford assay to normalize luciferase activity with protein present in the lysates. Then the relative luciferase activity was calculated according to control cells treated with DMSO set to 1. All experiments were performed in triplicates and the mean values with + SD are presented here.
Cell cycle analysis of PANC-1 cells treated with GSK3P inhibitors:
To determine effect of GSK3P inhibitors on cell death and/or cell cycle progression, PANC-1 cells were treated with identified molecules with or without TRAIL. Upon treatment with compounds in combination with TRAIL, PANC-1 cell death was determined with propidium iodide (PI) uptake measured by Flow cytometry. Treated cells were harvested in cold PBS and fixed in 70% ethanol at- 20 °C for overnight. The fixed cells were by centrifugation and re-suspended in PI (50 mg/mL) solution containing RNase (0.1 mg/mL) and Triton X-100 (0.05%). The cells were incubated in dark for for 1 h at room temperature. The excess PI was washed out using PBS, the resultant PI uptake was analyzed by FACS Caliber System (BD Bio- science, Erembodegem, Belgium) in an FL-2 fluorescence detector (10000 events were recorded for each condition). Flow cytometry data was analyzed using FCS express 4 software (De Novo Software, Los Angeles, California, USA).
Determination of caspase-3 and -9 activities in PANC-1 cells treated with GSK3P inhibitors: Both Caspase-3 and -9 activities were measured 48 h after treating the PANC-1 cells with either GSK3P inhibitors alone or in combination with TRAIL. Specific fluorogenic substrates Ac-DEVD-AMC and Ac-LEHD-AFC, were used specific to Caspase-3 and -9 respectively. The harvested cells were directly lysed in caspase lysis buffer (50 mM HEPES, 5mM CHAPS, 5 mM DTT, pH 7.5). The lysates were mixed with respective substrates prepared standard buffer (20 mM HEPES (pH 7.5), 0.1% CHAPS, 2 mM EDTA and 5 mM DTT) and incubated for 2 hrs at 37 °C in dark. The cleavage of substrates releasing AMC and AFC was detected by multimode reader (Perkin Elmer) using an excitation / emission wavelengths 380/460 nm for AMC and, 400 / 505 nm for AFC. The fluorescence readings were normalized to total protein concentration. The observed fluorescence is directly proportional to activity of caspase-3 and -9. The relative caspase activities were calculated as the ratio of values between DMSO treated and compound or standard drug treated cells.
Colony formation assay:
To determine the efficiency of GSK3P inhibitors in combination with the TRIAL on anchorage independent growth of PANC-1 cells soft agar assay were performed [3]. The base agar (0.5%) was prepared by mixing equal amounts of 1% agar with 2Xmedia (20% FBS and 2x antibiotics) filled in 6-well plate. Simultaneously, PANC-1 cells (14000/well) mixed with 2X media containing different concentrations of identified compounds alone or in combination with TRAIL mixed with 0.7% of sterile agar to achieve final concentration of 0.35% top agar. The cell suspension was spread on solidified base agar. Then the plates were incubated in C02 incubator for 14 days and the cells were fed with fresh media containing respective concentration and combination of TRIAL every 3 days. Upon termination (after 14 days of incubation) plates were stained with 0.005% of crystal violet for 1 h and excess stain was removed by washing with MQ. The number of blue colonies were counted using olympus Xi71 microscope.
Results and discussion:
Validation of docking pose:
The docking studies were concentrated in the specific ATP binding site, which is located between the N-terminal β-barrel domain and the C-terminal helical domain. The results suggest that three compounds are binding in the ATP binding site of GSK3P (Fig A) which showed interaction with the key residue VAL135. The compound C was interacting with ATP binding site of GSK3P beta pleated sheets. From the results it was observed that the presence of intermolecular H-bonding network between the CO and NH of the indole ring to the backbone NH of V135 and CO of D133 respectively. The binding energy is significantly low with score -9.2 Kcal/mol. Further the docking results from the compound C displayed similar binding mode with the presence of H-bonding network between CO of indole to NH of V135 and NH of Indole to CO of D133 with favourable docking score -7.5 Kcal/mol. For the compound U the H bond interaction was observed between the CO and NH of the indole ring to the backbone NH of V135 and NH of N65 with a favourable docking score -7.07 Kcal/mol. From the results, it is inferred that three compounds were interacting with specific ATP binding region and hinge residue V135 of GSK3P with favourable docking score and showed the better activity of the compounds. The docking poses were generated through PyMOL and the complex of three compounds interacting with binding site was pictorially represented in Figure A.
Spirooxindoles inhibits GSK3P kinase activity:
In invitro kinase assays screening of the small molecules archived in a compound library against GSK3P kinase activity has identified three potential fragments with IC50 values below 10 μΜ (Table 01). The library of spirooxindoles prepared in the present study was evaluated for their anti-kinase activity of GSK3p. The result indicates that ten compounds have potential to inhibit enzyme activity with IC50 <10 μΜ (Table 01). Though the compounds do not inhibit enzyme activity equivalent to commercially available inhibitors, observed IC50 values are better than LiCl standard GSK3P inhibitor.
Activity against intracellular GSK3P:
GSK3P is a part of the canonical β-catenin/Wnt pathway involved in promoting cell proliferation. GSK3P phosphorylates β-catenin targeting it for proteasomal degradation. Upon GSK3P inhibition, unphosphorylated β-catenin (active form) is translocated into nucleus, which then binds to tcf and lef family transcription factors and initiates transcription of target genes. Based on this principle we have established stable reporter cell line to monitor activation of wnt pathway. To determine intracellular efficiency of GSK3P inhibition, HEK293T cells (stably expressing tandem repeats of the TCF/LEF transcriptional response element followed by the firefly lucif erase) treated with selected compounds. The increased luciferase activity is a measure of activation of Wnt pathway (downstream of GSK3P) confirmed that the three compounds potentially inhibiting GSKP in a dose dependent manner (Figure 2A). GSKP usually inhibits the activity of its downstream target by protein phosphorylation of a protein onto threonine and serine amino acids residues. The immediate target of GSK3P is glycogen synthase (GS) which is involved in glyocgen synthesis there by regulating energy metabolosm [4] (Frame and Cohen, 2001). To confirm specificity of molecules inhibiting GSK3P, we have determined phosphorylation of glycogen synthase and accumulation of β-catenin. In the HEK293T cells treated with test compounds pGS levels are decreased and also increased levels of β-catenin confirms its stabilization due to decreased rate of Proteasomal degradation (Figure 2B). In the drug discovery pipeline, many of the newer molecules identified in target based approach often fail while hit to lead progression due to cell permeability. Therefore it is important to select molecules which can easily permeable through cell wall. The results obtained here confirmed that the selected compounds already fulfilled some important criteria such as, cell permeability and good inhibitory potential against endogenous GSKp.
Pancreatic cancer cells have already been reported to express high active pool of GSK3P [5]. Recent studies have revealed that the pancreatic cancer cells are resistant to TRAIL-induced apoptosis [6]. GSK3P inhibition sensitizes pancreatic and prostate cancer cells to TRAIL mediated apoptosis [7,8]. To determine efficiency of the identified molecules against proliferation of cancer cells, PANC-1 cells were treated with different concentrations of the identified compounds alone or in combination with TRAIL (10 ng/mL). The cytotoxicity results indicate that the compounds significantly increased sensitivity of PANC-1 cells to TRAIL (Figure 3 A). Among the three potential compounds tested AB-0 and AB-U inhibited the cell growth by 50% at 30 μΜ in combination with TRAIL (10 ng/mL) where as AB-C is less effective compared to other molecules. Higher concentration of test compounds AB-0 and AB-U showed more than 50% growth inhibition even at low concentration of TRAIL (1 ng/mL). The compound AB-C is not effective compared to remaining compounds (Figure 3B). TRAIL alone (10 ng/ml) did not cause any significant cell death in PANC-1 cells but TRAIL combination with GSK3P inhibitors in increasing concentrations cause significant decrease in cell viability.
To determine the long-term effects of the identified compounds against GSK3P on proliferation and anchorage independent growth of the PANC-1 cells soft agar assay (clonogenic assay) was performed. The cells were treated and fed continuously with compounds alone or along 10 ng/mL of TRAIL. Results clearly showed effect of AB-0 and AB-U on formation of colonies by PANC-1 cells. From the clonogenic assays, a dose- dependent inhibition was observed on treatment of cells with AB-0 and AB-U. Interestingly, both the compounds showed more effective inhibition of colony formation in the presence of TRAIL confirmed by size and morphology of the clones observed. The TRAIL alone did not show effective inhibition on colony formation at the concentration 10 ng/mL used (Figure 4). It has been reported that the clonogenic assay correlates well with in vivo assays of tumorigenicity in nude mice [9] indicating the high efficacy of AB-0 and AB-U towards inhibition of PANC-1 cells growth.
Further to confirm of cancer cells death by GSK3P inhibitors in combination with TRAIL, we have analyzed cell cycle progression of proliferating PANC-1 cells treated with different concentrations of compounds in the presence or absence of TRAIL. The histograms of cell cycle progression presented in the supplementary figure 01 and 02 suggest that both the compounds AB-0 and AB-U induces more cell death in combination with TRAIL than compounds alone or TRIAL alone. The increase in the % of cells in subGl indicates PANC-1 cells death. These results are in agrement with the previously reported results that the GSK3P inhibitors sensitize cancer cells to TRAIL induced cell death. TRAIL induced apoptosis of cancer cell is mediated by the activation of procaspases to active caspases by proteolysis. To glean more information weather the compounds show synergistic effect on activation of caspase cascade to induce apoptosis, we have measured caspase-3 and -9 activities in PANC- 1 cells treated with compounds along with TRAIL. The obtained result suggests that the combinatorial treatment of PANC- 1 cells is more effective even at lower concentration of the compounds than individually in activation of both the caspases (Figure 5). Taken together it is also clear that the apoptosis of PANC-1 cells treated with GSK3P inhibitors is through mitochondiral dependant pathway.
GSK3P is a multifunctional enzyme which phosphorylates several of its downstream substrates. GSK3P is actively involved in multiple signaling pathways thereby regulation of cell survival, proliferation, migration, inflammation, immune responses glucose regulation and insulin signaling, neuronal function and oncogenesis to embryonic development. Of the several substrates, some of the key targets are glycogen synthase, beta catenin and tau proteins [10,11]. To confirm potential of AB-0 and AB-U in combination with TRAIL on cellular GSK3P signalling, first we have measured phosphorylation of GSK3p. Increased phosphorylation of GSK3P on 9th serine residue silences its kinase activity (Figure 6A&B). In our results we observe the synergistic effect of both AB-0 and AB-U in combination with TRAIL in GSK3P inhibition by phosphorylation. Phosphorylation of GSK3P at serine residue will create a pseudo substrate conformation which folds and bound to the catalytic site of the enzyme and prevents binding of substrates to active site, there silences the GSK3P activity. Further, we measured phosphorylation of GS a primary substrate of GSK3P which is involved in glycogen metabolism. Here, p-GS was significantly decreased with increasing concentrations of AB-0 and AB-U, where total GS remains unaffected. The compounds are more effective when the PANC-lcells are treated in combination with TRAIL (Figure 6A&B).
GSKP participates in regulation of wnt pathway via phosphorylation of β-catenin. Active GSK3P phosphorylates β-catenin targeting it for ubiquitin mediated proteolysis. Inhibition of GSK3P causes the stabilization and accumulation of β-catenin in cytoplasm and further translocates into the nucleus. Nuclear β catenin along with TCF/LEF elements continuously drive transcription of its target genes. Nuclear β-catenin is implicated in progression of several types of cancers such as prostate cancer, basal cell carcinoma, head and neck cancers and medulloblastoma. As anticipated, β-catenin levels are elevated in PANC-1 cells exposed to GSK^ inhibitors compared to the control (DMSO) treated cells (Figure 6A&B). Interestingly, the synergistic effect of GSK^ inhibitors along with TRAIL on β-catenin levels is very significant compared to molecules or TRAIL alone.
Effect of GSK3P inhibition on NFkB transcriptional activity:
Daniel D. Billadeau et al reported that active GSK^ arbitrate cell proliferation and viability of the pancreatic cancer cells through NFkB pathway. NFkB is constitutively activated in most of the cancers including pancreatic cancer enhancing transcription of several genes responsible for cell survival [5] . To determine whether the compounds AB-0 and AB-U are NFkB dependent or independent in controlling growth of PANC-1 cells we have performed NFkB specific reporter assays. As showed in figure 05, lucif erase activity that is a measure of NFkB activity is significantly decreased with dose dependent inhibition of GSK3P using AB-0 and AB-U (Figure 7). Both the standard controls used also showed same effect in the PANC-1 cells.
Effect of AB-0 and AB-U on expression of NFkB target genes involved in cell survival: In cell viability assay we have observed significant reduction in cell viability upon the GSK3P inhibition by AB-0 and AB-U in combination with TRAIL. In NFkB reporter assays we have observed that the inhibition of GSK3P reduces NFkB activity. Altered transcriptional activity of NFkB may result in differential expression of its targeted genes such as anti-apoptotic proteins BCL2 and XIAP. Based on this result, we have measured the expression antiapoptotic proteins BCL2 and XIAP upon the inhibition of GSK3p. As showed in figure 8 A&B, we can clearly observe a significant decrease in the BCL2 and XIAP levels of PANC-1 cell treated with inhibitors. The compounds AB-0 and AB-U clearly showed synergistic effect on the BCL2 and XIAP levels. In presence of TRAIL both the compounds lowered expression of BCL2 and XIAP more effectively than TRAIL alone or compound alone. Shadi Mamaghani et al reported that cells over expressing the BCL2 an antiapoptotic protein are resistant to TRAIL induced apoptosis. Down regulation of BCL2 upon the GSK3P inhibition may sensitize the cells to TRAIL.
Further, we have measured expression of the Cyclin Dl in PANC-1 cells upon GSK3P inhibition. Both the compounds AB-0 and AB-U significantly reduced Cyclin Dl levels in combination with TRAIL (Figure 8 A&B). Cyclin Dl is a cell cycle regulating protein involved in control of cancer cell proliferation. Taken together, the results obtained indicate that the molecules AB-0 and AB-U inhibiting cellular GSK3P sensitizing the PANC-1 to TRAIL inducing apoptosis.
Discussion:
Glycogen Synthase Kinase 3β (GSK3P) is a serine/threonine kinase. It is a multi-functional enzyme, regulating several proteins involved in various signalling pathways including transcription factors, cell cycle/survival regulators and proto-oncogenes. Recent studies suggest that GSK3P may promote turn oogenesis in many cancer types. Its abberent over expression was found in human pancreatic (H), colon(I), prostate(J), leukaemia (K), thyroid(l), brain(M) and ovarian(N) cancer cells. Billadeau et. al. reported for elevated levels of GSK3P in many pancreatic cancer cell lines including PANC-1. In ovarian cancer patients GSK3P is significantly over expressed and active GSK3P promotes proliferation of ovarian cancer cells (N). This led to the current study design aiming to identify new GSK3P selective inhibitors and its inhibition is an attractive approach for effective treatment of pancreatic cancer.
From the series of synthesized compounds, AB-O, AB-C and AB-U showed potent inhibition of GSK3P in in- vitro kinase assays. To determine their efficiency against intracellular GSK3p; we observed that the treatment of PANC-1 cells with AB-O, AB-C and AB-U significantly decreased phosphorylation of GSK3p. Further, accumulation of β-catenin in these cells confirmed that these three molecules were effective against GSK3P in PANC-1 cells. Cell viability assays have confirmed that AB-O and AB-U are more effective in inhibiting cell proliferation. These results are in line with the previous studies that the inhibition of GSK3P decreases rate of cell proliferation and survival in many cancers such as CLL (k), pancreatic (h), colon (i), ovarian (n), thyroid (1) and brain (m).
Chemotherapeutic drugs are also known to be toxic to normal healthy cells, which is why it has become prerequisite in present time to develop effective strategies that increase the target based therapeutic potential of anticancer drugs with decreased toxicity to normal healthy cells, more efforts are being directed towards combination chemotherapy. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is known to induce apoptosis in various human cancer cell lines but not in normal healthy cells. Prostate and pancreatic cancer cell were resistant to TRIAL. David J. McConkey et al were measured the TRIAL dependent apoptosis in panel of 9 pancreatic cancer cells among them PANC-1 was shown to be less sensitivity to trail mediated apoptosis. in our study we measured growth inhibitory effects of new GSK3 inhibitors in combination with trail (10 ng/mL), based on our data there was significant decrease in cell viability was observed in combination with TRAIL.
GSK3 also plays a role in cell proliferation and apoptosis through several pathways. GSK regulates the various transcription factors which initiate the transcription of several proto oncogens. Present reports revealing that GSK3 positively regulates NFkB -dependent genes responsible for proliferation and anti-apoptotic signals in many cancers like pancreatic(d), and leukemia(K). In order to conform the above findings we measured the NFKB activity upon GSK3 inhibition, as we presented in figure 8 NFKB activity was increased. Further we have measured the expression of NFKB target genes whose were involved in apoptosis and cell proliferation. Both genes bcl2 and xiap were decreased in a dose dependent manner of AB-O and AB-U. Cyclin-dl, cell cycle regulator which promotes cell proliferation was also reduced. Recent studies showing pancreatic cancer cells were resistance to TRAIL, because of the increased expression of an anti-apoptotic protein XIAP. Inhibition of XIAP, sensitizes the pancreatic cancer cells to TRIAL mediated apoptosis in invitro and in vivo. There were many studies supporting the role of NFKB in trail resistance. It was explained by nfkb positively regulates the transcription of anti-apoptotic proteins such as XIAP, BCL-2, BCL- XL, and MCL-1 in cancer cells GSK mediates binding NFKB P65 subunit to the promoters of an anti-apoptotic genes BCL2 and XIAP, so inhibition of GSK reduces the expression these anti-apoptotic genes, there by inhibition of GSK sensitize the pancreatic cancer cells to trail mediated apoptosis.
To conform whether AB-0 and AB-U cause cell death in pancreatic cancer cells, we measured the cell death using FACS. The results indicate, panc-1 cells were showing cell death even at 25 uM concentration of AB-0 ,AB-U and death was increased with increasing doses of GSK inhibitors and in combination with the trail. Further to investigate, whether the cell death is because of apoptosis, we have measured the activity of caspases post exposure GSK inhibitors, both caspase3 and 9 were activated. Next we measured long term effects of GSK inhibitors in combination with the trail on panc-1 pancreatic cancer cells. The results showed that GSK inhibition reduces the anchorage of independent growth cancer cells this as an important future of cancer cells. Finally our data supports the anti-apoptotic role of GSK in pancreatic cancer, combination of GSK inhibition in combination with TRAIL or other drugs like gemcitabine may serve as chemotherapeutic agents to treat pancreatic cancer.
ADVANTAGES OF THE INVENTION
The advantages of the method are given below.
• The main advantage of the present invention is that it provides spirooxindole derivatives.
The advantage of the present invention is that it provides an efficient process for the preparation of diversely substituted novel spirooxindole derivatives.
Another advantage of the present invention is the use of these spirooxindole compounds as GSK3P inhibitors.
The spirooxindole compounds prepared are novel synthetic derivatives, which are useful as GSK3P inhibitors for their anticancer potential.

Claims

WE CLAIM
1. A Spirooxindole compound of formula I
Figure imgf000039_0001
Formula I
wherein
Z is hydrogen, halogen, hydroxy, alkoxy, cyano, nitro;
Ring A is a five membered or six membered or seven membered optionally containing additional hetero atom in the ring
X is optionally CH2 or none
Ring B is triazole or cyclic amine alone or with one or more hetero atoms part of the ring and optionally substituted with one or more substituents or no B ring present Y is hydrogen, halogen, azide, alkoxy, aryloxy, hydroxy, ester, amide, amino, alkyl or aryl, heteroaryl, wherein these substituents are optionally substituted with one or more halogen, hydroxy, alkoxy, alkyl, aryl, heteroaryl, aryloxy, nitro, cyano, ester, aldehyde,
wherein these substituents are further substituted with one or more halogen, hydroxy, alkoxy, alkyl, aryl, heteroaryl, aryloxy, nitro, cyano, ester, aldehyde.
2. The compounds as claimed in claim 1, wherein the spirooxindole compounds of formula I is represented by the compounds of formula II (9a)
Figure imgf000039_0002
Formula II wherein
R is hydrogen, alkoxy, aryloxy, hydroxy, ester, amide, amino, alkyl or aryl, heteroaryl, wherein these substituents are optionally substituted with one or more halogen, hydroxy, alkoxy, alkyl, aryl, heteroaryl, aryloxy, nitro, cyano, ester, aldehyde,
wherein these substituents are further substituted with one or more halogen, hydroxy, alkoxy, alkyl, aryl, heteroaryl, aryloxy, nitro, cyano, ester, aldehyde
4. The compounds as claimed in claim 1 and claim 2, wherein the spirooxindole compounds are represented by
(AB-O) Ethyl 2-methyl-6-(2-((l-((2'-oxo-4,5-dihydro-2H-spiro[furan-3,3'-indoline]- 5-yl)methyl)-lH-l,2,3-triazol-4-yl)methoxy)phenyl)nicotinate
(AB-M) Ethyl 6-(5-fluoro-2-((l-((2'-oxo-4,5-dihydro-2H-spiro[furan-3,3'-indoline]-5- yl)methyl)-lH-l,2,3-triazol-4-yl)methoxy)phenyl)-2-methylnicotinate
(AB-N) Methyl-6-(5-chloro-2-((l-((2'-oxo-4,5-dihydro-2H-spiro[furan-3,3'-indoline]-
5-yl)methyl)-lH-l,2,3-triazol-4-yl)methoxy)phenyl)-2-methylnicotinate
(AB-P) 2-((l-((2'-Oxo-4,5-dihydro-2H-spiro[furan-3,3'-indoline]-5-yl)methyl)-lH- l,2,3-triazol-4-yl)methoxy)benzaldehyde
(AB-W) Ethyl6-(3,5-dichloro-2-((l-((2'-oxo-4,5-dihydro-2H-spiro[furan-3,3'- indoline]-5-yl)methyl)-lH-l,2,3-triazol-4-yl)methoxy)phenyl)-2-methylnicotinate (AB-F) Ethyl-l-((2'-oxo-4,5-dihydro-2H-spiro[furan-3,3'-indoline]-5-yl)methyl)-lH- 1 ,2,3-triazole-4-carboxylate
(AB-C) 5-((4-Phenyl-lH-l,2,3-triazol-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'- indolin]-2'-one
(AB-G) 5-((4-(Hydroxymethyl)-lH-l,2,3-triazol-l-yl)methyl)-4,5-dihydro-2H- spiro [furan-3 ,3 '-indolin] -2'-one
(AB-E) 5-((4-(2-Hydroxypropan-2-yl)-lH-l,2,3-triazol-l-yl)methyl)-4,5-dihydro-2H- spiro [furan-3 ,3 '-indolin] -2'-one
(AB-D) 5-((4-(4-Nitrophenyl)-lH-l,2,3-triazol-l-yl)methyl)-4,5-dihydro-2H- spiro [furan-3 ,3 '-indolin] -2'-one
5. The compounds as claimed in claim 1 and claim 2, wherein the structural formulae of the representative compounds are
Figure imgf000041_0001
6. The compounds as claimed in claim 1, wherein the spirooxindole compounds of formula I is represented by the compounds of formula III (8)
Figure imgf000041_0002
8
Formula III
wherein
The ring B is cyclic amine alone or with one or more hetero atoms part of the ring and substituted with one or more substituents
R is hydrogen, alkoxy, aryloxy, hydroxy, ester, amide, amino, alkyl or aryl, heteroaryl, wherein these substituents are optionally substituted with one or more halogen, hydroxy, alkoxy, alkyl, aryl, heteroaryl, aryloxy, nitro, cyano, ester, aldehyde, wherein these substituents are further substituted with one or more halogen, hydroxy, alkoxy, alkyl, aryl, heteroaryl, aryloxy, nitro, cyano, ester, aldehyde.
7. The compounds as claimed in claim 1 and claim 5, wherein the spirooxindole compounds are represented by
(AB-X) 5-((4-Phenylpiperazin-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]- 2'-one
(AB-K) 5-(Morpholinomethyl)-4, 5-dihydro-2H-spiro [furan-3, 3' -indolin] -2'-one (AB-S) 5-((4-(Pyridin-2-yl)piperazin-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'- indolin]-2'-one
(AB-U) 5-((4-(2-Methoxyphenyl)piperazin-l-yl)methyl)-4,5-dihydro-2H-spiro[furan- 3 ,3 '-indolin] -2'-one
(AB-T) 5-((4-(4-Methoxyphenyl)piperazin-l-yl)methyl)-4,5-dihydro-2H-spiro[furan- 3 ,3 '-indolin] -2'-one
(AB-Q) 5-((4-Benzoylpiperazin-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'- indolin]-2'-one
(AB-L) 5-(Thiomorpholinomethyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]-2'-one (AB-R) 5-((4-Benzylpiperazin-l-yl)methyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]- 2'-one
(AB-V) 5-((4-(Bis(4-fluorophenyl)methyl)piperazin-l-yl)methyl)-4,5-dihydro-2H- spiro [furan-3 ,3 '-indolin] -2'-one
8. The compounds as claimed in claim 1 and claim 5, wherein the structural formulae of the re resentative compounds are
Figure imgf000042_0001
9. The compounds as claimed in claim 1, wherein the spirooxindole compounds of formula I is represented by the com ounds of formula IV (9b)
Figure imgf000043_0001
Formula IV
wherein
R = hydrogen, alkoxy, aryloxy, hydroxy, ester, amide, amino, alkyl or aryl, heteroaryl, wherein these substituents are optionally substituted with one or more halogen, hydroxy, alkoxy, alkyl, aryl, heteroaryl, aryloxy, nitro, cyano, ester, aldehyde,
wherein these substituents are further substituted with one or more halogen, hydroxy, alkoxy, alkyl, aryl, heteroaryl, aryloxy, nitro, cyano, ester, aldehyde.
10. The compounds as claimed in claim 1 and claim 8, wherein the spirooxindole compounds are represented by
(AC-A) Ethyl 6-(5-fluoro-2-((l-(2-oxo-2', 4', 5', 6'-tetrahydrospiro[indoline-3,3'- pyran]-5'-yl)-lH-l,2,3-triazol-4-yl)methoxy)-phenyl)-2-methylnicotinate
(AC-B) Ethyl 2-methyl-6-(2-((l-(2-oxo-2',4',5',6'-tetrahydrospiro[indoline-3,3'- pyran]-5'-yl)-lH-l,2,3-triazol-4-yl)methoxy)-phenyl)nicotinate
(AC-C) Ethyl 6-(5-chloro-2-((l-(2-oxo-2',4',5',6'-tetrahydrospiro [indoline-3,3'- pyran]-5'-yl)-lH-l,2,3-triazol-4- yl)methoxy) -phenyl)- 2-methyl nicotinate
(AC-D) Ethyl 6-(3,5-dichloro-2-((l-(2-oxo-2',4',5',6'-tetrahydrospiro-[indoline-3,3'- pyran]-5'-yl)-lH-l,2,3-triazol-4-yl)methoxy)phenyl)-2-methyl nicotinate
(AC-E) 2-((l-(2-Oxo-2', 4', 5', 6'-tetrahydrospiro[indoline-3,3'-pyran]-5'-yl)-lH-l,2,3- triazol-4-yl) methoxy)benzaldehyde
(AC-F) 5'-(4-(Pyridin-2-yl)-lH-l,2,3-triazol-l-yl)-2',4',5',6'-tetrahydrospiro[indoline- 3,3'-pyran]-2-one
(AC-G) 5'-(4-(Hydroxy(phenyl)methyl)-lH-l,2,3-triazol-l-yl)-2',4',5',6'-tetrahydro spiro[indoline-3,3'-pyran]-2-one
(AC-H) 5'-(4-((2-Nitrophenoxy)methyl)-lH-l,2,3-triazol-l-yl)-2',4',5',6'-tetrahydro spiro - [indoline-3 ,3 '-pyran] -2-one
(AC-I) 5'-(4-((4-Chloro-2-methylphenoxy)methyl)-lH-l,2,3-triazol-l-yl)-2',4',5',6'- tetra hydrospiro[indoline-3,3'-pyran]-2-one
(AC-J) 5'-(4-((3-Hydroxyphenylamino)methyl)-lH-l,2,3-triazol-l-yl)-2',4',5',6'- tetrahydrospiro[indoline-3,3'-pyran]-2-one
(AC-K) 5'-(4-((3-Aminophenoxy)methyl)-lH-l,2,3-triazol-l-yl)-2',4',5',6'-tetrahydro spiro[indoline-3,3'-pyran]-2-one
(AC-L) 5'-(4-((5-Bromo-3-nitropyridin-2-yloxy) methyl)-lH-l,2,3-triazol-l-yl)- 2',4',5',6'-tetrahydrospiro[indoline-3,3'-pyran]-2-one
11. The compounds as claimed in claim 1 and claim 8, wherein the structural formulae of the representative compounds are
Figure imgf000044_0001
AC-J AC-K AC-L
12. The compounds as claimed in claim 1, wherein the spirooxindole compounds of formula I is represented by the compounds of formula 6a, 6b, 7a, 7b
Figure imgf000045_0001
6a 6b 7a 7b
13. The compounds as claimed in claim 1 and claim 11, wherein the spirooxindole compounds are represented by
(6a) 5-(Iodomethyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]-2'-one
(6b) 5'-Iodo-2', 4', 5', 6'-tetrahydrospiro [indoline-3,3'-pyran]-2-one
(7a) 5-(Azidomethyl)-4,5-dihydro-2H-spiro[furan-3,3'-indolin]-2'-one
(7b) 5'-Azido-2',4',5',6'-tetrahydrospiro[indoline-3,3'-pyran]-2-one
14. A process for the preparation of compound of formula I as claimed in claim 1 comprising the steps of:
a) allylation of oxindole;
b) protection of free amine with boc group;
c) formylation using formaldehyde;
d) removal of the protection to give free amine;
e) cyclisation using iodine to give spiro-iodo compounds;
f) azidation of spiro-iodo compounds using sodium azide to give spiro-azide compounds or reaction of spiro-iodo compounds with cyclic amines to give the cyclic amine based spirooxindoles;
g) reaction of spiro-azide compounds with alkynes to give triazole based spirooxindoles under click chemistry conditions.
15. The compounds as claimed in claim 1, wherein the said compounds are screened for GSK3p activity. 16. The compounds as claimed in claim 1 and claim 15, wherein the said compounds for use as potential anticanceragent.
17. The compounds as claimed in claim 1 and claim 15, wherein the said compounds are useful as GSK3P inhibitors.
PCT/IN2018/050134 2017-03-10 2018-03-09 Spirooxindole compounds as gsk3β inhibitors and process for preparation thereof WO2018163216A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP18715984.3A EP3592747A1 (en) 2017-03-10 2018-03-09 Spirooxindole compounds as gsk3 inhibitors and process for preparation thereof
US16/492,359 US11306100B2 (en) 2017-03-10 2018-03-09 Spirooxindole compounds as GSK3B inhibitors and process for preparation thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN201611008487 2017-03-10
IN201611008487 2017-03-10

Publications (2)

Publication Number Publication Date
WO2018163216A1 true WO2018163216A1 (en) 2018-09-13
WO2018163216A9 WO2018163216A9 (en) 2019-08-22

Family

ID=61906800

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IN2018/050134 WO2018163216A1 (en) 2017-03-10 2018-03-09 Spirooxindole compounds as gsk3β inhibitors and process for preparation thereof

Country Status (3)

Country Link
US (1) US11306100B2 (en)
EP (1) EP3592747A1 (en)
WO (1) WO2018163216A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020163812A1 (en) 2019-02-08 2020-08-13 Frequency Therapeutics, Inc. Valproic acid compounds and wnt agonists for treating ear disorders
CN114478558A (en) * 2022-01-24 2022-05-13 郑州大学 Enantiomer-pure tetrahydrofuran spiro-oxoindole derivative and preparation method and application thereof

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0278352A2 (en) * 1987-02-10 1988-08-17 Bayer Ag Aminomethyl-tetrahydro-furanes
US6358750B1 (en) 1997-09-08 2002-03-19 Arqule, Inc. Spiro[pyrrolidine-2,3′-oxindole] compounds and method of use
US6774132B1 (en) 1999-07-21 2004-08-10 Astrazeneca Ab Spirooxindole derivatives that act as analgesics
EP1557417A1 (en) * 2003-12-19 2005-07-27 Sanofi-Aventis Substituted 8'-pyri(mi)dinyl-dihydrospiro-[cycloalkylamine]-pyrimido[1,2a] pyrimidin-6-one derivatives
WO2006110917A2 (en) 2005-04-11 2006-10-19 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their uses as therapeutic agents
WO2009073620A2 (en) * 2007-11-30 2009-06-11 Newlink Genetics Ido inhibitors
US20100041900A1 (en) * 2008-08-13 2010-02-18 Kuang Ta Foods Co., Ltd Isolation of lactam compound from adlay bran and its use on anti-proliferative cancer cells
WO2010045251A2 (en) 2008-10-17 2010-04-22 Xenon Pharmaceuticals, Inc. Spiro-oxindole compounds and their use as therapeutic agents
US20100105745A1 (en) * 2005-01-04 2010-04-29 Toshinari Minamoto Suppression of cancer and method for evaluating anticancer agent based on the effect of inhibiting gsk3 beta
WO2011047174A1 (en) 2009-10-14 2011-04-21 Xenon Pharmaceuticals Inc. Synthetic methods for spiro-oxindole compounds
US8101647B2 (en) 2008-10-17 2012-01-24 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their use as therapeutic agents
WO2014058035A1 (en) 2012-10-12 2014-04-17 Okinawa Institute Of Science And Technology School Corporation Novel spirooxindole derivative and process for producing the same
US8962828B2 (en) 2010-12-21 2015-02-24 Bayer Intellectual Property Gmbh Method for producing triazinyl-substituted oxindoles

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8735558B2 (en) 2005-02-14 2014-05-27 Pacific Arrow Limited Blocking the migration or metastasis of cancer cells by affecting adhesion proteins and the uses of new compounds thereof
AR077252A1 (en) 2009-06-29 2011-08-10 Xenon Pharmaceuticals Inc ESPIROOXINDOL COMPOUND ENANTIOMERS AND THEIR USES AS THERAPEUTIC AGENTS

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0278352A2 (en) * 1987-02-10 1988-08-17 Bayer Ag Aminomethyl-tetrahydro-furanes
US6358750B1 (en) 1997-09-08 2002-03-19 Arqule, Inc. Spiro[pyrrolidine-2,3′-oxindole] compounds and method of use
US6774132B1 (en) 1999-07-21 2004-08-10 Astrazeneca Ab Spirooxindole derivatives that act as analgesics
EP1557417A1 (en) * 2003-12-19 2005-07-27 Sanofi-Aventis Substituted 8'-pyri(mi)dinyl-dihydrospiro-[cycloalkylamine]-pyrimido[1,2a] pyrimidin-6-one derivatives
US20100105745A1 (en) * 2005-01-04 2010-04-29 Toshinari Minamoto Suppression of cancer and method for evaluating anticancer agent based on the effect of inhibiting gsk3 beta
WO2006110917A2 (en) 2005-04-11 2006-10-19 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their uses as therapeutic agents
WO2009073620A2 (en) * 2007-11-30 2009-06-11 Newlink Genetics Ido inhibitors
US20100041900A1 (en) * 2008-08-13 2010-02-18 Kuang Ta Foods Co., Ltd Isolation of lactam compound from adlay bran and its use on anti-proliferative cancer cells
WO2010045251A2 (en) 2008-10-17 2010-04-22 Xenon Pharmaceuticals, Inc. Spiro-oxindole compounds and their use as therapeutic agents
US8101647B2 (en) 2008-10-17 2012-01-24 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their use as therapeutic agents
WO2011047174A1 (en) 2009-10-14 2011-04-21 Xenon Pharmaceuticals Inc. Synthetic methods for spiro-oxindole compounds
EP2488531A1 (en) 2009-10-14 2012-08-22 Xenon Pharmaceuticals Inc. Synthetic methods for spiro-oxindole compounds
US8742109B2 (en) 2009-10-14 2014-06-03 Xenon Pharmaceuticals Inc. Synthetic methods for spiro-oxindole compounds
US8962828B2 (en) 2010-12-21 2015-02-24 Bayer Intellectual Property Gmbh Method for producing triazinyl-substituted oxindoles
WO2014058035A1 (en) 2012-10-12 2014-04-17 Okinawa Institute Of Science And Technology School Corporation Novel spirooxindole derivative and process for producing the same

Non-Patent Citations (17)

* Cited by examiner, † Cited by third party
Title
ADV. SYNTH. CATAL., vol. 355, 2013, pages 1023
ANGEW. CHEM., vol. 116, 2004, pages 2719
BIOORG. MED. CHEM. LETT., vol. 16, 2006, pages 2105
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 13 May 2015 (2015-05-13), XP002781174, Database accession no. 1702718-68-0 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 24 May 2015 (2015-05-24), XP002781177, Database accession no. 1691090-70-6 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 26 January 2017 (2017-01-26), XP002781171, Database accession no. 2059927-67-0 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 27 January 2017 (2017-01-27), XP002781170, Database accession no. 2060027-64-5 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 29 April 2015 (2015-04-29), XP002781176, Database accession no. 1694142-26-1 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 6 May 2015 (2015-05-06), XP002781175, Database accession no. 1699579-55-9 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 8 August 2016 (2016-08-08), XP002781173, Database accession no. 1969003-24-4 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 9 September 2016 (2016-09-09), XP002781172, Database accession no. 1989963-10-1 *
IOANNIS A. KATSOULIS ET AL: "Synthesis of 5,6-Spiroethers and Evaluation of their Affinities for the Bacterial A Site", CHEMBIOCHEM - A EUROPEAN JOURNAL OF CHEMICAL BIOLOGY., vol. 12, no. 8, 6 May 2011 (2011-05-06), DE, pages 1188 - 1192, XP055475822, ISSN: 1439-4227, DOI: 10.1002/cbic.201100076 *
J. MED. CHEM., vol. 19, 1976, pages 892
J. MED. CHEM., vol. 49, 2006, pages 3432
LUYI ZHU ET AL: "Enantioselective Construction of Spirocyclic Oxindole Derivatives with Multiple Stereocenters via an Organocatalytic Michael/Aldol/Hemiacetalization Cascade Reaction", ORGANIC LETTERS , 14(23), 6012-6015 CODEN: ORLEF7; ISSN: 1523-7052, vol. 18, no. 10, 20 May 2016 (2016-05-20), pages 2387 - 2390, XP055476211, ISSN: 1523-7060, DOI: 10.1021/acs.orglett.6b00873 *
ORG. LETT., vol. 13, 2011, pages 3086
SHASHWAT MALHOTRA ET AL: "Design, synthesis and biological activity evaluation of regioisomeric spiro-(indoline-isoxazolidines) in the inhibition of TNF-[alpha]-induced ICAM-1 expression on human endothelial cells", MEDCHEMCOMM, vol. 3, no. 12, 1 January 2012 (2012-01-01), United Kingdom, pages 1536, XP055476213, ISSN: 2040-2503, DOI: 10.1039/c2md20216f *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020163812A1 (en) 2019-02-08 2020-08-13 Frequency Therapeutics, Inc. Valproic acid compounds and wnt agonists for treating ear disorders
CN114478558A (en) * 2022-01-24 2022-05-13 郑州大学 Enantiomer-pure tetrahydrofuran spiro-oxoindole derivative and preparation method and application thereof

Also Published As

Publication number Publication date
US20210284650A1 (en) 2021-09-16
WO2018163216A9 (en) 2019-08-22
US11306100B2 (en) 2022-04-19
EP3592747A1 (en) 2020-01-15

Similar Documents

Publication Publication Date Title
JP6750806B2 (en) Substituted heterocyclyl derivatives as CDK inhibitors
CN103588771B (en) There is the miazines antineoplastic compound of activity of hedgehog path antagonist
Liu et al. Senolytic activity of piperlongumine analogues: synthesis and biological evaluation
Liu et al. Design, synthesis and structure-activity relationships of novel 4-phenoxyquinoline derivatives containing 1, 2, 4-triazolone moiety as c-Met kinase inhibitors
HRP20031081A2 (en) Cdk inhibiting pyrimidines, production thereof and their use as medicaments
CN108774214A (en) Heterocycleamide as kinase inhibitor
KR20150112953A (en) Teatrahydro- and dihydro-isoquinoline prmt5 inhibitors and uses thereof
CN104936951A (en) Novel benzimidazole derivatives as kinase inhibitors
Malebari et al. β-Lactams with antiproliferative and antiapoptotic activity in breast and chemoresistant colon cancer cells
EA023176B1 (en) Arylsulfonyl pyrazoline carboxamidine derivatives as 5-htantagonists
CN102803246A (en) HEDGEHOG pathway antagonists and therapeutic applications thereof
US20200291011A1 (en) Coumarin derivatives, processes for their preparation and uses thereof for the treatment of cancer
Hou et al. Structure-based discovery of 1H-indole-2-carboxamide derivatives as potent ASK1 inhibitors for potential treatment of ulcerative colitis
US11306100B2 (en) Spirooxindole compounds as GSK3B inhibitors and process for preparation thereof
Zheng et al. Design, synthesis, biological activity evaluation of 3-(4-phenyl-1H-imidazol-2-yl)-1H-pyrazole derivatives as potent JAK 2/3 and aurora A/B kinases multi-targeted inhibitors
Hopkins et al. Optimization of novel reversible Bruton’s tyrosine kinase inhibitors identified using tethering-fragment-based screens
Zhou et al. N-Arylsulfonylsubstituted-1H indole derivatives as small molecule dual inhibitors of signal transducer and activator of transcription 3 (STAT3) and tubulin
Li et al. Semisynthetic derivatives of fradcarbazole A and their cytotoxicity against acute myeloid leukemia cell lines
Song et al. Discovery of bazedoxifene analogues targeting glycoprotein 130
CN109641909A (en) The mechanism target and its treatment use of rapamycin signal pathway inhibitor
CN106946896B (en) Furans simultaneously [2,3-d] pyrimidine -4- amine derivative
CN101514179A (en) Indolone hydrazine ethylidene carbonyl compound, method for preparing same and application thereof in medicine
Kandukuri et al. Design and Synthesis of Some New Quinoxaline Containing 1, 3, 4-Oxadiazole Hybrids and Evaluation of Their Anti-Cancer Activity
Wang et al. Design, synthesis and anti-tumor efficacy of novel phenyl thiazole/triazole derivatives as selective TrkA inhibitors
VM Sharma et al. Synthesis and biological evaluation of triazole-vanillin molecular hybrids as anti-cancer agents

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18715984

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2018715984

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2018715984

Country of ref document: EP

Effective date: 20191010